Language selection

Search

Patent 3057157 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3057157
(54) English Title: BIOMARKERS FOR CANCER THERAPEUTICS
(54) French Title: BIOMARQUEURS POUR THERAPIES ANTICANCEREUSES
Status: Deemed Abandoned
Bibliographic Data
(51) International Patent Classification (IPC):
  • G01N 33/50 (2006.01)
  • A61K 35/763 (2015.01)
  • A61K 39/395 (2006.01)
  • G01N 33/574 (2006.01)
  • G01N 33/68 (2006.01)
(72) Inventors :
  • GANSERT, JENNIFER LORRAINE (United States of America)
  • ANDERSON, ABRAHAM ANTONIO (United States of America)
  • GORSKI, KEVIN (United States of America)
(73) Owners :
  • MERCK SHARP & DOHME CORP.
  • AMGEN INC.
(71) Applicants :
  • MERCK SHARP & DOHME CORP. (United States of America)
  • AMGEN INC. (United States of America)
(74) Agent: BORDEN LADNER GERVAIS LLP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2018-04-27
(87) Open to Public Inspection: 2018-11-01
Examination requested: 2022-09-26
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/US2018/029915
(87) International Publication Number: WO 2018201028
(85) National Entry: 2019-09-18

(30) Application Priority Data:
Application No. Country/Territory Date
62/491,746 (United States of America) 2017-04-28

Abstracts

English Abstract

Biomarkers useful for identifying a variety of cancers that are responsive to treatment with a combination therapy comprising pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof and talimogene laherparepvec are provided. Methods of treating cancers that are resistant to monotherapy with pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof are provided. Methods of treating a cancer in a subject having a tumor with a low CD8+ density, a low or negative interferon gamma signature, and/or a low or negative PD-L1 status are also provided.


French Abstract

L'invention concerne des biomarqueurs utiles pour identifier divers cancers qui sont sensibles au traitement par une polythérapie à base de pembrolizumab, d'un variant ou fragment de celui-ci se liant à l'antigène et de talimogène laherparepvec. L'invention concerne également des méthodes de traitement des cancers qui sont résistants à la monothérapie à base de pembrolizumab, d'un variant ou un fragment de celui-ci se liant à l'antigène. Des méthodes de traitement d'un cancer chez un sujet ayant une tumeur caractérisée par une faible densité de CD8+, une signature d'interféron gamma faible ou négative et/ou un état PD-L1 faible ou négatif sont en outre décrites.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
What is claimed is:
1. A method of treating a tumor in a subject comprising:
selecting a subject having a tumor comprising a CD8+ T-cell infiltration
density of
fewer than about 1500, about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells / mm2;
administering talimogene laherparepvec to the subject; and
administering pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment thereof to the subject.
2. The method of claim 1, wherein the tumor expresses lower levels of two,
three, four
or five interferon gamma (IFN.gamma.) signature genes prior to administering
compared to a pre-
specified threshold of a control panel of signature genes selected from the
group consisting of
IFN.gamma., signal transducer and activator of transcription 1 (STAT1), C-C
chemokine receptor
type 5 (CCR5), chemokine (C-X-C motif) ligand 9 (CXCL9), perforin 1 (PRF1),
BLA-DRA,
chemokine (C-X-C motif) ligand 10 (CXCL10), chemokine (C-X-C motif) ligand 11
(CXCL11), indoleamine 2,3-dioxygenase 1 (TDO1) and granzyme A (GZMA).
3. The method of claim 1, wherein the tumor expresses no IFNy signature
genes prior to
administering the talimogene laherparepvec and the pembrolizumab, the
pembrolizumab
variant or the antigen-binding fragment thereof.
4. The method of claim 1, wherein the tumor has a programmed death-ligand 1
(PD-L1)
status of less than about 50% prior to administering the talimogene
laherparepvec and the
pembrolizumab, the pembrolizumab variant or the antigen-binding fragment
thereof.
5. The method of claim 1, wherein the tumor has a PD-L1 status of less than
about 1%
prior to administering the talimogene laherparepvec and the pembrolizumab, the
pembrolizumab variant or the antigen-binding fragment thereof.
94

6. The method of claim 1, wherein the talimogene laherparepvec is
administered to the
subject prior to the administration of the pembrolizumab or the antigen-
binding fragment
thereof.
7. The method of claim 1, wherein the subject has a cancer selected from
the group
consisting of melanoma, non-small cell lung cancer, head and neck cancer,
colorectal cancer,
breast cancer, ovarian cancer, bladder cancer, prostate cancer, sarcoma, renal
cell cancer,
gastric cancer, esophageal cancer, anal canal cancer, biliary tract cancer and
pancreatic
cancer.
8. The method of claim 7, wherein:
the melanoma is cutaneous melanoma, metastatic melanoma, or uveal melanoma;
the breast cancer is HER2+ breast cancer, HER2- HR+ breast cancer, or triple-
negative breast cancer;
the prostate cancer is castration-resistant prostate cancer;
the bladder cancer is transitional cell cancer or urothelial cancer;
the head and neck cancer is reculTent or metastatic squamous cell carcinoma of
the
head and neck; and/or
the sarcoma is soft tissue sarcoma or bone sarcoma.
9. The method of claim 1, wherein the tumor comprises a CD8+ T-cell
infiltration
density of fewer than about 1000 cells / Mm2.
10. A method of treating a tumor in a subject that is poorly responsive to
or non-
responsive to monotherapy with pembrolizumab, a pembrolizumab variant or an
antigen-
binding fragment thereof comprising:
administering talimogene laherparepvec to the subject; and
administering pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment thereof to the subject.
11. The method of claim 10, wherein:

the tumor comprises a CD8+ T-cell infiltration density of fewer than about
1500,
about 1400, about 1300, about 1200, about 1100, about 1000, about 900, about
800, about
700, about 600, or about 500 cells / mm2prior to administering;
the tumor expresses lower levels of two, three, four or five interferon gamma
(IFN.gamma.)
signature genes prior to administering than a pre-specified threshold of a
control panel of
signature genes selected from the group consisting of IFNy, STAT1, CCR.5,
CXCL9, PRF1,
HLA-DRA, CXCLIO, CXCL11, IDO1 and GZMA; and/or
the tumor has a PD-L1 status of less than about 50% prior to administering the
talimogene laherparepvec and the pembrolizumab, the pembrolizumab variant or
the antigen-
binding fragment thereof.
12. A method of treating a tumor in a subject that progressed during
monotherapy with
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof,
comprising:
administering talimogene laherparepvec to the subject; and
administering pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment thereof to the subject.
13. The method of chum 12, wherein:
the tumor comprises a CDS+ T-cell infiltration density of fewer than about
1500,
about 1400, about 1300, about 1200, about 1100, about 1000, about 900, about
800, about
700, about 600, or about 500 cells / mm2prior to administering;
the tumor expresses lower levels of two, three, four or five interferon gamma
(IFNy)
signature genes prior to administering than a pre-specified threshold of a
control panel of
signature genes selected from the group consisting of IFNy, STAT1, CCR5,
CXCL9, PRF1,
HLA-DRA, CXCL10, CXCL11, IDO1 and GZMA; and/or
the tumor has a PD-L1 status of less than about 50% prior to administering the
talimogene laherparepvec and the pembrolizumab, the pembrolizumab variant or
the antigen-
binding fragment thereof.
14. A method of treating a tumor having a CD8 T-cell infiltration density
of fewer than
about 1500, about 1400, about 1300, about 1200, about 1100, about 1000, about
900, about
800, about 700, about 600, or about 500 cells / mm2 comprising contacting the
tumor with
96

talimogene laherparepvec and pembrolizumab, a pembrolizumab variant or an
antigen-
binding fragment thereof.
15. The method of claim 14, wherein the tumor is from a subject having a
cancer selected
from the group consisting of melanoma, non-small cell lung cancer, head and
neck cancer,
colorectal cancer, breast cancer, ovarian cancer, bladder cancer, prostate
cancer, sarcoma,
renal cell cancer, gastric cancer, esophageal cancer, anal canal cancer,
biliary tract cancer and
pancreatic cancer.
16. The method of claim 15, wherein:
the melanoma is cutaneous melanoma, metastatic melanoma, or uveal melanoma;
and/or
the head and Iva cancer is recurrent or metastatic squamous cell carcinoma of
the
head and neck.
17. The method of claim 14, wherein the tumor expresses lower levels of
two, three, four
or five interferon gamma (IFN.gamma.) signature genes prior to contacting than
a pre-specified
threshold of a control panel of signature genes selected from the group
consisting of IFNy,
STAT1, CCR5, CXCL9, PRF1, HLA-DRA, CXCL10, CXCL11, IDO1 and GZMA.
18. The method of claim 14, wherein the tumor has a PD-L1 of less than
about 50% prior
to contacting with the talimogene laherparepvec and the pembrolizumab, the
pembrolizumab
variant or the antigen-binding fragment thereof.
19. A method of treating a tumor expressing lower levels of two, three,
four or five
interferon gamma (IFNy) signature genes prior to treatment than a pre-
specified threshold of
a control panel of signature genes elected from the group consisting of IFNy,
STAT1, CCR5,
CXCL9, PRF1, HLA-DRA, CXCL10, CXCL11, IDO1 and GZMA, comprising contacting
the tumor with talimogene laherparepvec and pembrolizumab, a pembrolizumab
variant or an
antigen-binding fragment thereof.
20. The method of claim 19, wherein the tumor is from a subject having a
cancer selected
from the group consisting of melanoma, non-small cell lung cancer, head and
neck cancer,
97

colorectal cancer, breast cancer, ovarian cancer, bladder cancer. prostate
cancer, sarcoma,
renal cell cancer, gastric cancer. esophageal cancer, anal canal cancer,
biliary tract cancer and
pancreatic cancer.
21. The method of claim 20, wherein:
the melanoma is cutaneous melanoma, metastatic melanoma, or uveal melanoma;
and/or
the head and neck cancer is recurrent or metastatic squamous cell carcinoma of
the
head and neck.
22. The method of claim 19, wherein the tumor has a CD8 T-cell infiltration
density of
fewer than about 1500, about 1400, about 1300, about 1200, about 1100, about
1000. about
900, about 800, about 700, about 600, or about 500 cells / mm2prior to
contacting.
23. The method of claim 19, wherein the tumor has a PD-L1 status of less
than about 50%
prior to contacting with the talimogene laherparepvec and the pembrolizumab, a
pembrolizumab variant or an antigen-binding fragment thereof.
24. A method of treating a tumor having a PD-L1 status of less than about
50% prior to
treatment, comprising contacting the tumor with talimogene laherparepvec and
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof.
25. The method of claim 24, wherein the tumor is from a subject having a
cancer selected
from the group consisting of melanoma, non-small cell lung cancer, head and
neck cancer,
colorectal cancer, breast cancer, ovarian cancer, bladder cancer, prostate
cancer, sarcoma,
renal cell cancer, gastric cancer, esophageal cancer, anal canal cancer,
biliary tract cancer and
pancreatic cancer.
26. The method of claim 25, wherein:
the melanoma is cutaneous melanoma, metastatic melanoma, or uveal melanoma;
and/or
the head and neck cancer is recurrent or metastatic squamous cell carcinoma of
the
head and neck.
98

27. The method of claim 24, wherein the tumor has a CD8+ T-cell
infiltration density of
fewer than about 1500, about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells / mm2 prior to
contacting.
28. The method of claim 24, wherein the tumor expresses lower levels of
two, three, four
or five interferon gamma (IFN.gamma.) signature genes prior to contacting than
a pre-specified
threshold of a control panel of signature genes selected from the group
consisting of IFN.gamma.,
STAT1, CCR5, CXCL9, PRF1, HLA-DRA, CXCL10, CXCL11, IDO1 and GZMA.
29. A method of treating a tumor having a CD8 T-cell infiltration density
of fewer than
about 1500, about 1400, about 1300, about 1200, about 1100, about 1000, about
900, about
800, about 700, about 600, or about 500 cells / mm2 and expressing lower
levels of five or
fewer interferon gamma (IFN.gamma.) signature genes prior to treatment than a
pre-specified
threshold of a control panel of signature genes selected from the group
consisting of IFN.gamma.,
STAT1, CCR5, CXCL9, PRF1, HLA-DRA, CXCL10, CXCL11, IDO1 and GZMA,
comprising contacting the tumor with talimogene laherparepvec and
pembrolizumab, a
pembrolizumab variant or an-antigen-binding fragment thereof.
30. The method of claim 29, wherein the tumor is from a subject having a
cancer selected
from the group consisting of melanoma, non-small cell lung cancer, head and
neck cancer,
colorectal cancer, breast cancer, ovarian cancer, bladder cancer, prostate
cancer, sarcoma,
renal cell cancer, gastric cancer, esophageal cancer, anal canal cancer,
biliary tract cancer and
pancreatic cancer.
31. The method of claim 30, wherein:
the melanoma is cutaneous melanoma, metastatic melanoma, or uveal melanoma;
and/or
the head and neck cancer is recurrent or metastatic squamous cell carcinoma of
the
head and neck.
99

32. The method of claim 29, wherein the tumor has a PD-L1 status of less
than about 50%
prior to contacting with the talimogene laherparepvec and the pembrolizumab, a
pembrolizumab variant or an antigen-binding fragment thereof.
33. A method of treating a previously pembrolizumab-, pembrolizumab variant-
or
antigen-binding fragment thereof-resistant rumor in a subject subsequently
exposed to
talimogene laherparepvec comprising administering to the subject
pembrolizumab, a
pembrolizumab variant or an antigen-binding fragment thereof.
34. The method of claim 33, wherein the subject has a cancer selected from
the group
consisting of melanoma, non-small cell lung cancer, head and neck cancer,
colorectal cancer,
breast cancer, ovarian cancer, bladder cancer, prostate cancer, sarcoma, renal
cell cancer,
gastric cancer, esophageal cancer, anal canal cancer, biliary tract cancer and
pancreatic
cancer.
35. The method of claim 34, wherein:
the melanoma is cutaneous melanoma, metastatic melanoma, or uveal melanoma;
and/or
the head and neck cancer is recurrent or metastatic squamous cell carcinoma of
the
head and neck.
36. A method of rendering a tumor that is resistant to monotherapy with
pembrolizumab,
a pembrolizumab variant or an antigen-binding fragment thereof in a subject
sensitive to
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof,
comprising
contacting the tumor with talimogene laherparepvec.
37. The method of claim 36, wherein a sample of the tumor taken from the
subject after
contacting the tumor with talimogene laherparepvec has an increased level of
one or any
combination of CD8+ T-cells, CD4+ T-cells, IFN.gamma., CD20+ B-cells, memory T-
cells,
regulatory T-cells and CD56+ cells relative to a sample of the tumor taken
prior to the
contacting the tumor with talimogene laherparepvec.
100

38. The method of claim 36, wherein the tumor has a CD8+ T-cell
infiltration density of
greater than 1000 cells / mm2 after contacting with talimogene laherparepvec.
39. A method of treating a tumor in a subject comprising:
selecting a subject having a tumor comprising a CD8+ T-cell infiltration
density of
fewer than about 1500, about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells / mm2;
administering talimogene laherparepvec to the subject intratumorally as an
initial dose
followed by one or more secondary doses; and
administering pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment thereof to the subject systemically as an initial dose followed by
one or more
secondary doses.
101

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
BIOMARKERS FOR CANCER THERAPEUTICS
RELATED APPLICATIONS
[001] This application claims the benefit of priority of U.S. Provisional
Application No.
62/491,746, filed April 28, 2017. The contents of the aforementioned
application is hereby
incorporated by reference in its entirety for all purposes.
FIELD OF THE INVENTION
[002] The present invention relates to the field of cancer therapeutics. In
particular, the
present invention relates to biomarkers useful for identifying a variety of
cancers that can be
treated with a combination therapy comprising pembrolizumab, a pembrolizumab
variant
and/or an antigen-binding fragment thereof and talimogene laherparepvec.
BACKGROUND
[003] Treatment with anti-PD-1 or anti-PD-Li antibodies results in long
lasting anti-tumor
responses in patients with a variety of cancers, and it is becoming standard
of care treatment
for patients with metastatic melanoma, carcinomas of the head and neck, lung,
kidney and
bladder, as well as Merkel cell carcinoma and Hodgkin's disease (Sharma, P.,
and Allison,
J.P. (2015). The future of immune checkpoint therapy. Science 348, 56-61).
However, in all
of these indications, only a subset of patients respond to therapy, with a
majority of patients
being primarily resistant to PD-1 blockade. Accordingly, new cancer treatments
targeting
PD-1 blockade-resistant cancers are needed.
SUMMARY
[004] The present disclosure is based on the discovery of biomarkers (e.g.,
intratumoral
biomarkers) that can be used to identify a tumor that is responsive to
combination therapy
with pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof and
talimogene laherparepvec. The present invention is particularly useful for
treating tumors in
subjects that were previously untreatable or not sufficiently treatable with
monotherapy (i.e.,
with a checkpoint inhibitor (e.g., with anti-PD-L1 therapy or anti-PD-1
therapy (e.g.,
1

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)).
Intratumoral administration of the oncolytic virus talimogene laherparepvec (a
herpes
simplex virus type 1 designed to preferentially replicate in tumors and
produce granulocyte¨
macrophage colony-stimulating factor (GM-CSF)) was determined to increase
intratumoral
infiltration by cytotoxic T-cells in human patients, thereby improving the
anti-tumor activity
of the anti-PD-1 antibody pembrolizumab when used as a combination therapy in
subjects
with tumors that exhibit a low CD8+ T-cell density, a low or negative
interferon gamma
signature, and/or a low or negative PD-L1 status, and/or in subjects that were
non-responsive
or poorly responsive to previous checkpoint inhibitor therapy (e.g., with anti-
PD-Li therapy
or anti-PD-1 therapy (e.g., pembrolizumab), or are unlikely to respond to
monotherapy with a
checkpoint inhibitor (e.g., with anti-PD-L1 therapy or anti-PD-1 therapy
(e.g.,
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)) due to,
e.g., low PD-L1 status.
[005] In one aspect, a method of treating a tumor in a subject comprising
selecting a subject
having a tumor comprising a CD8+ T-cell infiltration density of fewer than
about 1500, about
1400, about 1300, about 1200, about 1100, about 1000, about 900, about 800,
about 700,
about 600, or about 500 cells / mm2, administering talimogene laherparepvec to
the subject,
and administering pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment
thereof, to the subject is provided.
[006] In certain embodiments, the tumor expresses lower levels of two, three,
four or five
interferon gamma (IFNy) signature genes prior to administering compared to a
pre-specified
threshold of a control panel of signature genes selected from the group
consisting of IFNy,
signal transducer and activator of transcription 1 (STAT1), C-C chemokine
receptor type 5
(CCR5), chemokine (C-X-C motif) ligand 9 (CXCL9), perforin 1 (PRF1), HLA-DRA,
chemokine (C-X-C motif) ligand 10 (CXCL10), chemokine (C-X-C motif) ligand 11
(CXCL11), indolefunine 2,3-dioxygenase 1 (11)01) and granzyme A (GZMA). In
certain
embodiments, the tumor expresses no IFNy signature genes prior to
administering the
talimogene laherparepvec and the pembrolizumab, the pembrolizumab variant or
the antigen-
binding fragment thereof.
[007] In certain embodiments, the tumor has a programmed death-ligand 1 (PD-
L1) status
of less than about 50% prior to administering the talimogene laherparepvec and
the
pembrolizumab, the pembrolizumab variant or the antigen-binding fragment
thereof. In
certain embodiments, the tumor has a PD-Li status of less than about 1% prior
to
2

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
administering the talimogene laherparepvec and the pembrolizumab, the
pembrolizumab
variant or the antigen-binding fragment thereof.
[008] in certain embodiments, the talimogene laherparepvec is administered to
the subject
intratumorally and/or the pembrolizumab or the antigen-binding fragment
thereof is
administered to the subject systemically.
[009] In certain embodiments, the talimogene laherparepvec is administered to
the subject
prior to the administration of the pembrolizumab or the antigen-binding
fragment thereof.
[010] In certain embodiments, a reduction in size of the injected tumor and/or
a reduction in
size of a non-injected tumor occur after administering the talimogene
laherparepvec and the
pembrolizumab, the pembrolizumab variant or the antigen-binding fragment
thereof.
[011] In certain embodiments, CD8+ T-cell infiltration density is increased in
the tumor
after administering the talimogene laherparepvec. In certain embodiments,
dividing CD8+ T-
cells circulating in the subject are increased after administering the
talimogene laherparepvec
and the pembrolizumab, the pembrolizumab variant or the antigen-binding
fragment thereof.
[012] In certain embodiments, the subject has a cancer selected from the group
consisting of
melanoma, non-small cell lung cancer, head and neck cancer, colorectal cancer,
breast
cancer, ovarian cancer, bladder cancer, prostate cancer, sarcoma, renal cell
cancer, gastric
cancer, esophageal cancer, anal canal cancer, biliary tract cancer and
pancreatic cancer. In
certain embodiments, the melanoma is cutaneous melanoma, metastatic melanoma,
or uveal
melanoma, the breast cancer is HER2+ breast cancer, HER2- HR+ breast cancer,
or triple-
negative breast cancer, the prostate cancer is castration-resistant prostate
cancer, the bladder
cancer is transitional cell cancer or urothelial cancer, the head and neck
cancer is recurrent or
metastatic squamous cell carcinoma of the head and neck, and/or the sarcoma is
soft tissue
sarcoma or bone sarcoma.
[013] In certain embodiments, the tumor comprises a CD8+ T-cell infiltration
density of
fewer than about 1000 cells / mm2.
[014] In one aspect, a method of treating a tumor in a subject that is poorly
responsive to or
non-responsive to monotherapy with a checkpoint inhibitor (e.g., with anti-PD-
Li therapy or
anti-PD-1 therapy (e.g., pembrolizumab, a pembrolizumab variant or an antigen-
binding
fragment thereof)) comprising administering talimogene laherparepvec to the
subject, and
administering pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment
thereof to the subject is provided.
3

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[015] In certain embodiments, the tumor comprises a CD8+ T-cell infiltration
density of
fewer than about 1500, about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells / mm2 prior to
administering, the
tumor expresses lower levels of two, three, four or five interferon gamma
(IFNy) signature
genes prior to administering than a pre-specified threshold of a control panel
of signature
genes selected from the group consisting of IFNy, STAT1, CCR5, CXCL9, PRF1,
HLA-
DRA, CXCL10, CXCL11, IDO1 and GZMA, and/or the tumor has a PD-L1 status of
less
than about 50% prior to administering the talimogene laherparepvec and the
pembrolizumab,
the pembrolizumab variant or the antigen-binding fragment thereof.
[016] In one aspect, a method of treating a tumor in a subject that progressed
during
monotherapy with a checkpoint inhibitor (e.g., with anti-PD-L1 therapy or anti-
PD-1 therapy
(e.g., pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)),
comprising administering talimogene laherparepvec to the subject, and
administering
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof
to the
subject is provided.
[017] In certain embodiments, the tumor comprises a CD8+ T-cell infiltration
density of
fewer than about 1500, about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells / mm2 prior to
administering, the
tumor expresses lower levels of two, three, four or five 1FNy signature genes
prior to
administering than a pre-specified threshold of a control panel of signature
genes selected
from the group consisting of IFNy, STAT1, CCR5, CXCL9, PRF1, HLA-DRA, CXCL10,
CXCL11, IDO1 and GZMA, and/or the tumor has a PD-L1 status of less than about
50%
prior to administering the talimogene laherparepvec and the pembrolizumab, the
pembrolizumab variant or the antigen-binding fragment thereof.
[018] In certain embodiments, the talimogene laherparepvec is administered
sequentially as
an initial dose followed by one or more secondary doses. In certain
embodiments, the
pembrolizumab, the pembrolizumab variant or the antigen-binding fragment
thereof is
administered sequentially as an initial dose followed by one or more secondary
doses. In
certain embodiments, the talimogene laherparepvec is administered sequentially
as an initial
dose followed by one or more secondary doses, and the pembrolizumab, the
pembrolizumab
variant or the antigen-binding fragment thereof is administered sequentially
and
concomitantly with the one or more secondary doses of the talimogene
laherparepvec.
4

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[019] In certain embodiments, the talimogene laherparepvec is administered
intratumorally
and wherein the pembrolizumab, the pembrolizumab variant or the antigen-
binding fragment
thereof is administered systemically. In certain embodiments, the talimogene
laherparepvec
and the pembrolizumab, the pembrolizumab variant or the antigen-binding
fragment thereof,
are administered intratu morally.
[020] In one aspect, a method of treating a tumor having a CD8+ T-cell
infiltration density
of fewer than about 1500, about 1400, about 1300, about 1200, about 1100,
about 1000,
about 900. about 800. about 700, about 600, or about 500 cells / mm2
comprising contacting
the tumor with talimogene laherparepvec and pembrolizumab, a pembrolizumab
variant or an
antigen-binding fragment thereof is provided.
[021] In certain embodiments, the tumor is from a subject having a cancer
selected from the
group consisting of melanoma, non-small cell lung cancer, head and neck
cancer. colorectal
cancer, breast cancer, ovarian cancer, bladder cancer, prostate cancer,
sarcoma, renal cell
cancer, gastric cancer, esophageal cancer, anal canal cancer, biliary tract
cancer and
pancreatic cancer. In certain embodiments, the cancer is cutaneous melanoma.
In certain
embodiments, the cancer is recurrent or metastatic squamous cell carcinoma of
the head and
neck.
[022] In certain embodiments, the tumor expresses lower levels of two, three,
four or five
1FNy signature genes prior to contacting than a pre-specified threshold of a
control panel of
signature genes selected from the group consisting of IFNy, STAT1, CCR5,
CXCL9, PRF1,
HLA-DRA. CXCLIO. CXCLII, IDO1 and GZMA.
[023] In certain embodiments, the tumor has a PD-Li of less than about 50%
prior to
contacting with the talimogene laherparepvec and the pembrolizumab, the
pembrolizumab
variant or the antigen-binding fragment thereof.
[024] In one aspect, a method of treating a tumor expressing lower levels of
two, three, four
or five IFNy signature genes prior to treatment than a pre-specified threshold
of a control
panel of signature genes elected from the group consisting of IFNy, STAT1,
CCR5, CXCL9,
PRF1, HLA-DRA, CXCL10, CXCL11, IDO1 and GZMA, comprising contacting the tumor
with talimogene laherparepvec and pembrolizumab, a pembrolizumab variant or an
antigen-
binding fragment thereof is provided.
[025] in certain embodiments, the tumor is from a subject having a cancer
selected from the
group consisting of melanoma, non-small cell lung cancer, head and neck
cancer, colorectal
cancer, breast cancer, ovarian cancer, bladder cancer. prostate cancer,
sarcoma, renal cell

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
cancer, gastric cancer, esophageal cancer, anal canal cancer, biliary tract
cancer and
pancreatic cancer. In certain embodiments, the cancer is melanoma (e.g.,
cutaneous
melanoma). In certain embodiments, the cancer is head and neck cancer (e.g.,
recurrent or
metastatic squamous cell carcinoma of the head and neck).
[026] In certain embodiments, the tumor has a CD8+ T-cell infiltration density
of fewer than
about 1500. about 1400, about 1300, about 1200, about 1100, about 1000, about
900. about
800, about 700, about 600, or about 500 cells / mm2 prior to contacting.
[0271 In certain embodiments, the tumor has a PD-L1 status of less than about
50% prior to
contacting with the talimogene laherparepvec and the pembrolizumab, a
pembrolizumab
variant or an antigen-binding fragment thereof.
[028] In one aspect, a method of treating a tumor having a PD-Li status of
less than about
50% prior to treatment, comprising contacting the tumor with talimogene
laherparepvec and
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof
is provided.
[029] In certain embodiments, the tumor is from a subject having a cancer
selected from the
group consisting of melanoma, non-small cell lung cancer, head and neck
cancer, colorectal
cancer, breast cancer, ovarian cancer, bladder cancer, prostate cancer,
sarcoma, renal cell
cancer, gastric cancer, esophageal cancer, anal canal cancer, biliary tract
cancer and
pancreatic cancer. In certain embodiments, the cancer is melanoma (e.g.,
cutaneous
melanoma). In certain embodiments. the cancer is head and neck cancer (e.g.,
recurrent or
metastatic squamous cell carcinoma of the head and neck).
[030] In certain embodiments, the tumor has a CD8+ T-cell infiltration density
of fewer than
about 1500, about 1400, about 1300, about 1200, about 1100. about 1000, about
900, about
800, about 700, about 600, or about 500 cells / mm2 prior to contacting.
[031] In certain embodiments, the tumor expresses lower levels of two, three,
four or five
IFNy signature genes prior to contacting than a pre-specified threshold of a
control panel of
signature genes selected from the group consisting of IFNy, STAT1, CCR5,
CXCL9, PRF1,
HLA-DRA. CXCLIO. CXCL11, IDO1 and GZMA.
[032] In one aspect, a method of treating a tumor having a CD8+ T-cell
infiltration density
of fewer than about 1500, about 1400, about 1300, about 1200, about 1100,
about 1000,
about 900. about 800, about 700, about 600, or about 500 cells / mm2 and
expressing lower
levels of five or fewer IFNy signature genes prior to treatment than a pre-
specified threshold
of a control panel of signature genes selected from the group consisting of
IFNy, STAT1,
CCR5, CXCL9, PRF1. HLA-DRA, CXCL10, CXCL11, IDO1 and GZMA, comprising
6

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
contacting the tumor with talimogene laherparepvec and pembrolizumab, a
pembrolizumab
variant or an-antigen-binding fragment thereof is provided.
[033] in certain embodiments, the tumor is from a subject having a cancer
selected from the
group consisting of melanoma, non-small cell lung cancer, head and neck
cancer, colorectal
cancer, breast cancer, ovarian cancer, bladder cancer, prostate cancer,
sarcoma, renal cell
cancer, gastric cancer, esophageal cancer, anal canal cancer, biliary tract
cancer and
pancreatic cancer. In certain embodiments, the cancer is melanoma (e.g.,
cutaneous
melanoma). In certain embodiments, the cancer is head and neck cancer (e.g.,
recurrent or
metastatic squamous cell carcinoma of the head and neck).
[034] in certain embodiments, the tumor has a PD-L1 status of less than about
50% prior to
contacting with the talimogene laherparepvec and the pembrolizumab, a
pembrolizumab
variant or an antigen-binding fragment thereof.
[035] In one aspect, a method of treating a previously pembrolizumab-,
pembrolizumab
variant- or antigen-binding fragment thereof-resistant tumor in a subject
subsequently
exposed to talimogene laherparepvec comprising administering to the subject
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof
is provided.
[036] In certain embodiments, the subject has a cancer selected from the group
consisting of
melanoma, non-small cell lung cancer, head and neck cancer, colorectal cancer,
breast
cancer, ovarian cancer, bladder cancer, prostate cancer, sarcoma, renal cell
cancer, gastric
cancer, esophageal cancer, anal canal cancer, biliary tract cancer and
pancreatic cancer. In
certain embodiments, the cancer is melanoma (e.g., cutaneous melanoma). In
certain
embodiments, the cancer is head and neck cancer (e.g., recurrent or metastatic
squamous cell
carcinoma of the head and neck).
[037] In one aspect, a method of rendering a tumor that is resistant to
monotherapy with a
checkpoint inhibitor (e.g., with anti-PD-L1 therapy or anti-PD-1 therapy
(e.g.,
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)) in a
subject sensitive to pembrolizumab, a pembrolizumab variant or an antigen-
binding fragment
thereof, comprising contacting the tumor with talimogene laherparepvec is
provided.
[038] In certain embodiments, a sample of the tumor taken from the subject
after contacting
the tumor with talimogene laherparepvec has an increased level of one or any
combination of
CD8+ T-cells, CD4+ T-cells, IFNy, CD20+ B-cells, memory T-cells, regulatory T-
cells and
CD56+ cells relative to a sample of the tumor taken prior to the contacting
the tumor with
talimogene laherparepvec.
7

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[039] In certain embodiments, the tumor has a CD8+ T-cell infiltration density
of greater
than 1000 cells / mm2 after contacting with talimogene laherparepvec.
[040] in certain embodiments, a blood sample taken from the subject after
contacting the
tumor with talimogene laherparepvec has an increased level of CD8+ T-cells
and/or CD4+ T-
cells compared to a blood sample taken from the subject prior to the
contacting, optionally
wherein the CD8+ T-cells are dividing CD8+ T-cells.
[041] In one aspect, a method of treating a tumor in a subject comprising
selecting a subject
having a tumor comprising a CD8+ T-cell infiltration density of fewer than
about 1500, about
1400, about 1300, about 1200, about 1100, about 1000, about 900, about 800,
about 700,
about 600, or about 500 cells / mm2, administering talimogene laherparepvec to
the subject
intratumorally as an initial dose followed by one or more secondary doses, and
administering
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof
to the
subject systemically as an initial dose followed by one or more secondary
doses is provided.
[042] In certain embodiments, the secondary doses are administered every two
weeks
(Q2W). In certain embodiments, the initial dose of talimogene laherparepvec is
administered
on day 1 of week 1 and a secondary dose of talimogene laherparepvec is
administered on day
1 of week 4, on day 1 of week 6, and Q2W thereafter. In certain embodiments,
the initial
dose of pembrolizumab, pembrolizumab variant or antigen-binding fragment
thereof is
administered on day 1 of week 6 and a secondary dose of pembrolizumab,
pembrolizumab
variant or antigen-binding fragment thereof is administered on day 1 of week 8
and Q2W
thereafter.
[043] In certain embodiments, the initial dose of talimogene laherparepvec is
administered
at a dose of 106 plaque forming units (pfu)/mL and the secondary doses of
talimogene
laherparepvec are administered at a dose of 108 pfu/mL.
[044] In certain embodiments, the initial dose of pembrolizumab, pembrolizumab
variant or
antigen-binding fragment thereof is administered at a dose of 200 mg and the
secondary
doses of pembrolizumab, pembrolizumab variant or antigen-binding fragment
thereof are
administered at a dose of 200 mg.
[045] The summary of the disclosure described above is non-limiting and other
features and
advantages of the disclosed biomarkers and methods will be apparent from the
following
drawings, detailed description of the disclosure, and claims.
8

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
BRIEF DESCRIPTION OF THE DRAWINGS
[046] Figure 1 depicts a melanoma study design and clinical response to a
combination of
talimogene laherparepvec and pembrolizumab. (A) Sets forth the phase lb study
design
schema. Stars indicate the time of scheduled tumor biopsies. (B) Depicts
computer
tomographic scans of two patients with response to the combination therapy.
Melanoma
metastases are marked with a blue arrow at baseline. (C) Depicts a waterfall
plot of best
response change in tumor burden from baseline. Patients were required to have
baseline and
>1 post-baseline tumor assessments to be included. (D) Depicts a change in
tumor burden
over time. (E) Depicts a Kaplan-Meier analysis of progression-free survival.
(F) Depicts a
Kaplan-Meier analysis of overall survival.
[047] Figure 2 illustrates that a combination of talimogene laherparepvec and
pembrolizumab was effective in patients with low tumor CD8 density. (A)
Depicts baseline
CD8 density in tumor biopsies according to response rate. The magnitude of
bars indicates
baseline tumor CD8 density in each patient's baseline biopsy, and best overall
response is
indicated on x-axis and by bar color. Red: complete response (CR); pink:
partial response
(PR), black: progressive disease (PD). (B) Baseline PD-L1 by IHC status (1%
cut off) and
interferon gamma signature score by NanoString analysis is shown under each
patient's CD8
result. Best overall response per investigator is shown as of cutoff date of
August 2016. n.a.
= result not available.
[048] Figure 3 shows that talimogene laherparepvec increased tumor CD8 density
in
patients responding to a combination of talimogene laherparepvec and
pembrolizumab. (A)
Depict examples of pre- (week 1) and post- (week 6) talimogene laherparepvec
and
talimogene laherparepvec plus pembrolizumab (week 30) CD8+ T-cell density in
tumor
biopsies: visualization of cells stained with CD8 antibody with red chromogen.
Staining was
quantified for tissue regions of interest including CD8 density in tumor as
shown for
talimogene laherparepvec injected tumors. (B) Depicts CD8 density, and (C)
granzyme-B H-
score which is shown for baseline and post baseline biopsies. The left side in
each panel
shows post baseline results from injected lesions and the right side in each
panel shows
results from non-injected lesions. Open circles indicate results from tumor
biopsies that were
depleted of melanoma cells, but that had pathological features of having
previously been
infiltrated by melanoma cells such as melanin deposits. Response is color
coded for best
overall response per investigator: complete or partial response in red and non-
response in
9

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
blue. (D) Depicts CD8a and (E) interferon gamma normalized mRNA transcript
count
measured in the NanoString Pan Cancer Immune Profiling Panel.
[049] Figure 4 shows that talimogene laherparepvec increases tumor
infiltrating
lymphocyte density and PD-L1 expression in tumors. Twelve color
immunofluorescence
staining was performed on a single slide from paired pre- and post-talimogene
laherparepvec
tumor biopsies from each of 13 patients. Markers evaluated included S100 (as
melanoma
segmentation marker), CD3, CD4, CD8, PD-1, PD-L1, CTLA-4, CD45RO, Foxp3, CD56,
CD68 and CD20. (A) Depicts a subset of changes at week 6 from baseline in
marker cell
positive cell density for results with statistical significance (PD-L1, PD-1,
CD8, CD4, CD56,
CD20, CD45R0 and Foxp3) are graphed for non-injected samples (left) and
injected samples
(right). Median change for each subset is shown with a horizontal line.
Response is color
coded for best overall response per investigator: complete or partial response
in red and non-
response in blue. (B) Shows an example of the combination of S100 (blue), CD8
(green) and
PD-Li (red) staining is shown at low (top row) and high (bottom row)
magnification for a
baseline biopsy from a patient who went onto have a partial response (week 1),
week 6 after
injection of talimogene laherparepvec, and at week 30 after long term
treatment with the
combination of talimogene laherparepvec and pembrolizumab. NI: biopsy of a non-
injected
metastasis; T: biopsy of an injected metastasis.
[050] Figure 5 depicts circulating T-cell subsets and expression of activation
markers.
Peripheral blood cells obtained from baseline, week 1, week 6, week 8, and
week 30 were
analyzed by flow cytometry. (A) Fold-change in absolute CD3+/CD8+ cells. (B)
Depicts
fold-change in absolute CD3+/CD4+ cells. (C) Depicts percent change in Ki67+
(CD3+/CD8+) cells. (D) Depicts percent change in PD-1+ (CD3+/CD8+) cells at
week 1 and
week 6 only, as after starting on pembrolizumab the staining antibody competed
for the same
epitope. (E) Depicts percent change in TIM3+ (CD3+/CD8+) cells. (F) Depicts
percent
change in BTLA+ (CD3+/CD8+) cells. P-values for comparison to baseline are
shown below
data for each post-baseline visit, based on contrasts from linear mixed
effects modeling.
Response is color coded for best overall response per investigator: complete
or partial
response in red and non-response in blue.
[051] Figure 6 depicts the disposition of patients enrolled in the study and
biopsy
availability for biomarker testing.

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[052] Figure 7 depicts changes in tumor burden at the lesion level. (A)
Depicts injected
lesion response. (B) Depicts non-injected, non-visceral lesion response. (C)
Depicts non-
injected, visceral lesion response.
[053] Figure 8 shows that responders had CD8 density increases in injected
lesions after
talimogene laherparepvec administration. Changes from baseline (BL, week 1) in
CD8
density as measured by IHC after talimogene laherparepvec treatment but before
the start of
combination therapy (W6) are plotted for responders and non-responders
separately. The left
panel shows changes in injected lesions (Inj) and the right panel shows
changes in non-
injected (Not Inj) lesions on a fold-change scale. Response was defined as
best overall
response per investigator of CR or PR, and non-response as PD or SD. Tumor-
depleted
samples are indicated with open circles. Median fold change is indicated with
a horizontal
line (solid for all samples, dashed for only those with tumor present).
[054] Figure 9 shows that talimogene laherparepvec decreases Treg fraction of
CD4 T-cells
in tumors. Twelve color immunofluorescence staining was performed on a single
slide from
paired tumor biopsies at pre- and post-talimogene laherparepvec from each of
13 patients.
Markers evaluated included S100 (as melanoma segmentation marker), CD3, CD4,
CD8, PD-
1, PD-L1, CTLA-4, CD45RO, Foxp3, CD56, CD68 and CD20. Changes from baseline in
Treg fraction of CD4 T-cells at week 6 are graphed for non-injected samples
(left) and
injected samples (right). Median change for each subset is shown with a
horizontal line.
Response is color-coded for best overall response per investigator: complete
or partial
response in red and non-response in blue.
[055] Figure 10 depicts additional multi-parameter imaging (MultiOmyxTm
platform)
examples from a patient with a partial response to therapy are shown for (A)
the combination
of S100, CD8 and PD-L1 (0.6 mm2 and 0.04 mm2 image area) at baseline (week 1),
week 6
after injection of talimogene laherparepvec, and at week 30 after long term
treatment with the
combination of talimogene laherparepvec and pembrolizumab. (B) Depicts S100,
CD3, CD4
and Foxp3 staining (0.6 mm2 and 0.04 mm2 image area) for an additional
patient.
[056] Figure 11 depicts an IFNI, gene signature panel according to certain
embodiments of
the invention.
[057] Figure 12 depicts a schematic of a phase 3 study design and treatment
schema in
which subjects are treated with talimogene laherparepvec plus pembrolizumab
(arm 1) or
placebo plus pembrolizumab (arm 2) until 24 months from the date of the first
dose of
pembrolizumab or until the end of treatment due to disappearance of injectable
lesions,
11

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
complete response, disease progression per irRC-RECIST or intolerance of study
treatment
(AE). AE, Adverse Event requiring permanent discontinuation of study
treatment; CR,
Complete Response; PD, Progressive Disease; Rx, treatment; T-VEC, talimogene
laherparepvec. 'Subjects will be followed-up for serious adverse events until
90 (+ 7) days
after the last dose of talimogene laherparepvec or the last dose of
pembrolizumab, whichever
is later. bLong-term follow-up will be performed every 12 weeks ( 28 days)
until
approximately 60 months after the last subject enrolled in phase 3.
DETAILED DESCRIPTION OF EXEMPLARY EMBODIMENTS
[058] It has been established that a high level of CD8+ T-cell
infiltration, a high level of
PD-Ll and/or a positive IFNy gene signature in a tumor are required for the
successful
treatment of a tumor using PD-1/PD-L1 antagonists. Indeed, current cancer
therapies that
target the programmed death-1 (PD-1) receptor have shown unprecedented rates
of durable
clinical responses in patients with various cancer types, including tumor
regression after
therapeutic PD-1 blockade, with such results requiring a high level of pre-
existing CD8+ T-
cells (Tumeh et al. (2014) Nature 515:568-571). An anti-PD-L1 antibody was
determined to
be efficacious in treating multiple cancer types in patients with tumors
expressing high levels
of PD-Li (Herbst et al. (2014) Nature 515:563-7). In addition, the presence of
a high
interferon gamma (IFNy) gene signature in a tumor correlates with the ability
to treat the
tumor with PD-1 antagonists (WO 2015/094992; Applicant, Merck Sharp & Dohme
Corporation).
[059] Surprisingly, and quite contrary to the accepted dogma that a high level
of CD8+ T-
cell infiltration, a high PD-Ll level, and/or a positive IFNy gene signature
is necessary to
successfully treat a tumor with a PD-1 antagonist, it has been discovered that
tumors having
low levels of CD8+ T-cell infiltration, a low or negative PD-Li status, and/or
a low or
negative 1FNy gene signature profile could be effectively treated with the PD-
1 antagonist
pembrolizumab using a combination therapy approach with talimogene
lahetparepvec. This
discovery represents the first clinical demonstration that talimogene
laherparepvec could
trigger an immune response in a tumor in vivo sufficient to enable efficient
targeting of the
tumor by the PD-1 antagonist pembrolizumab.
[060] it was further discovered that intratumoral administration of talimogene
laherparepvec
favorably altered the tumor microenvironment of injected lesions by, for
example, increasing
12

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
CD8+ T-cell infiltration, increasing PD-L1 expression, and/or producing a more
positive
IFNy gene signature, thus rendering tumor cells more susceptible to anti-PD-1
therapy.
Expression of PD-Li increased after treatment with talimogene laherparepvec
but was
countered by subsequent PD-1 blockade with pembrolizumab. After injection of
talimogene
laherparepvec into a lesion, tumor antigen-specific CD8+ T-cells trafficked to
and infiltrated
both the local lesion as well as distant metastatic lesions. Using a
combination therapy of
pembrolizumab and talimogene laherparepvec, an anti-PD-1 blockade acted to
counter
checkpoint protein-mediated inhibition of the immune response.
[061] Thus, administration of the oncolytic virus rendered "cold" tumors
(i.e., tumors
exhibiting a low level of immune infiltration (e.g., by CD8+ T cells), a
negative IFNy gene
signature, and/or a low PD-Li status) more susceptible to pembrolizumab
blockage therapy
by converting such tumors to "hot" tumors (i.e., tumors exhibiting high levels
of immune
infiltration (e.g., by CD8+ T cells), a positive IFNy gene signature, and/or a
high PD-Li
status). Accordingly, tumors that previously were minimally responsive to or
unresponsive to
monotherapy with a checkpoint inbibitor (e.g., with anti-PD-L1 therapy or anti-
PD-1 therapy
(e.g., pembrolizumab or a variant or antigen-binding fragment thereof)) could
be rendered
sensitive to therapy with pembrolizumab, or a variant or antigen-binding
fragment thereof.
[062] Accordingly, in one embodiment, the combination of pembrolizumab, a
pembrolizumab variant or an antigen-binding fragment thereof and talimogene
laherparepvec
is used to increase the magnitude of tumor specific T-cell responses as
compared to
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof
alone in
patients. This effect can be particularly observed with previously untreated,
unresectable,
stable 111b-IV melanoma. The combination of pembrolizumab, a pembrolizumab
variant or
an antigen-binding fragment thereof and talimogene laherparepvec is intended
to enhance the
systemic anti-tumor response to tumor antigens following the lytic replication
of talimogene
laherparepvec in tumors. Therefore, the combination therapy can result in
enhanced
destruction of injected tumors as well as uninjected/distant tumors, including
micro-
metastatic disease, to improve the rate of overall tumor response and duration
of response. In
total, these effects can contribute to an improvement in overall survival,
particularly when
compared to monotherapy with a checkpoint inbibitor (e.g., with anti-PD-Li
therapy or anti-
PD-1 therapy (e.g., pem.brolizumab, a pembrolizumab variant or an antigen-
binding fragment
thereof)). The use of talimogene laherparepvec in combination with
pembrolizumab, a
pembrolizumab variant or an antigen-binding fragment thereof, is intended to
enhance T-cell
13

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
activation through different mechanisms, respectively augmenting dendritic
cell-mediated
tumor antigen presentation (Kaufman et al., Ann Surg Oncol., 17(3):718-730,
2010)
following the release of tumor antigens by lytic virus replication, enhanced
through the local
expression of GM-CSF, and antagonizing immune tolerance by blocking inhibitory
signals
mediated by an immune checkpoint inhibitor (Kapadia and Fong, J Clin Oncol.,
23:8926-
8928, 2005).
[063] In certain embodiments, a cold tumor-associated cancer having CD8+ T-
cell
infiltration density of fewer than about 1500, about 1400, about 1300, about
1200, about
1100, about 1000, about 900, about 800, about 700, about 600, or about 500
cells per 1 nun2
sample (or per 1 mL sample), a low or negative PD-L1 status and/or a negative
IFNy gene
signature is treated with a combination of pembrolizumab, a pembrolizumab
variant or an
antigen-binding fragment thereof and talimogene laherparepvec. Such tumors
include, but
are not limited to, melanoma (e.g., cutaneous, metastatic, uveal), non-small
cell lung cancer,
head and neck cancer (e.g., recurrent or metastatic squamous cell carcinoma of
the head and
neck), colorectal cancer, breast cancer (e.g., HER2+, HER2- (11R+), triple-
negative), ovarian
cancer, bladder cancer (e.g., transitional cell cancer, urothelial cancer),
prostate cancer (e.g.,
castration-resistant), sarcoma (e.g., soft tissue, bone), renal cell cancer,
gastric cancer,
esophageal cancer, anal canal cancer, biliary tract cancer and pancreatic
cancer.
[064] In other embodiments, a hot tumor (or a cancer associated with a hot
tumor) (i.e., a
cancer that is not associated with one or more of a CD8+ T-cell infiltration
density of fewer
than about 1500, about 1400, about 1300, about 1200, about 1100, about 1000,
about 900,
about 800, about 700, about 600, or about 500 cells per 1 mrn2 sample (or per
1 mL sample),
a low or negative PD-L1 status and/or a low level of IFNy gene signature
expression) that is
responsive to monotherapy with a checkpoint inbibitor (e.g., with anti-PD-Li
therapy or anti-
PD-1 therapy (e.g., pembrolizumab, a pembrolizumab variant or an antigen-
binding fragment
thereof)) is treated with a combination of pembrolizumab, a pembrolizumab
variant or an
antigen-binding fragment thereof and talimogene laherparepvec. Such cancers
include, but
are not limited to, melanoma (e.g., cutaneous, metastatic, uveal), lung cancer
(e.g., non-small
cell lung cancer, small cell lung cancer), head and neck cancer (e.g.,
recurrent or metastatic
squamous cell carcinoma of the head and neck), nasopharyngeal cancer, thyroid
cancer,
salivary cancer, esophageal cancer), breast cancer (e.g., ER+/HER2- breast
cancer, triple-
negative breast cancer), ovarian cancer, cervical cancer, bladder cancer
(e.g., urothelial
cancer), renal cell cancer, gastrointestinal cancer (e.g., hepatocellular
cancer, colorectal
14

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
cancer, anal cancer), biliary tract cancer, multiple myeloma, lymphoma (e.g.,
mediastinal
large B-cell lymphoma, Hodgkin's lymphoma) and mesothelioma.
Definitions
[065] So that the invention may be more readily understood, certain technical
and scientific
terms are specifically defined below. Unless specifically defined elsewhere in
this document,
all other technical and scientific terms used herein have the meaning commonly
understood
by one of ordinary skill in the art to which this invention belongs.
[066] As used herein, including the appended claims, the singular forms of
words such as
"a," "an," and "the," include their corresponding plural references unless the
context clearly
dictates otherwise.
[067] "About" when used to modify a numerically defined parameter (e.g., the
gene
signature score for a gene signature discussed herein, or the dosage of a
pembrolizumab,
pembrolizumab variant and/or antigen-binding fragment thereof or talimogene
laherparepvec,
or the length of treatment time with a pembrolizumab, pembrolizumab variant
and/or antigen-
binding fragment thereof or talimogene laherparepvee) means that the parameter
may vary by
as much as 10% above or below the stated numerical value for that parameter.
For example,
a gene signature consisting of about 10 genes may have between 9 and 11 genes.
[068] "Administration" and "treatment," as it applies to an animal, human,
experimental
subject, cell, tissue, organ, or biological fluid, refers to contact of an
exogenous
pharmaceutical, therapeutic, diagnostic agent, or composition to the animal,
human, subject,
cell, tissue, organ, or biological fluid. Treatment of a cell encompasses
contact of a reagent
to the cell, as well as contact of a reagent to a fluid, where the fluid is in
contact with the cell.
"Administration" and "treatment" also means in vitro and ex vivo treatments,
e.g., of a cell,
by a reagent, diagnostic, binding compound, or by another cell.
[069] As used herein, the term "antibody" refers to any form of antibody that
exhibits the
desired biological or binding activity. Thus, it is used in the broadest sense
and specifically
covers, but is not limited to, monoclonal antibodies (including full-length
monoclonal
antibodies), polyclonal antibodies, multi-specific antibodies (e.g.,
bispecific antibodies),
humanized antibodies, fully human antibodies, chimeric antibodies and
camelized single
domain antibodies. "Parental antibodies" are antibodies obtained by exposure
of an immune
system to an antigen prior to modification of the antibodies for an intended
use, such as
humanization of an antibody for use as a human therapeutic.

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[070] In general, the basic antibody structural unit comprises a tetramer.
Each tetramer
includes two identical pairs of polypeptide chains, each pair having one
"light" (about 25
kDa) and one "heavy" chain (about 50-70 klla). The amino-terminal portion of
each chain
includes a variable region of about 100 to 110 or more amino acids primarily
responsible for
antigen recognition. The carboxy-terminal portion of the heavy chain may
define a constant
region primarily responsible for effector function. Typically, human light
chains are
classified as kappa and lambda light chains. Furthermore, human heavy chains
are typically
classified as mu, delta, gamma, alpha, or epsilon, and define the antibodys
isotype as IgM,
IgD, IgG, IgA, and IgE, respectively. Within light and heavy chains, the
variable and
constant regions are joined by a "J" region of about 12 or more amino acids,
with the heavy
chain also including a "D" region of about 10 more amino acids. See generally,
Fundamental
Immunology Ch. 7 (Paul, W., ed., 2nd ed. Raven Press, N.Y. (1989)).
[071] The variable regions of each light/heavy chain pair form the antibody
binding site.
Thus, in general, an intact antibody has two binding sites. Except in
bifunctional or
bispecific antibodies, the two binding sites are, in general, the same.
[072] Typically, the variable domains of both the heavy and light chains
comprise three
hypervariable regions, also called complementarity determining regions (CDRs),
which are
located within relatively conserved framework regions (FR). The CDRs are
usually aligned
by the framework regions, enabling binding to a specific epitope. In general,
from N-
terminal to C-terminal, both light and heavy chains variable domains comprise
FR1, CDR1,
FR2, CDR2, FR3, CDR3 and FR4. The assignment of amino acids to each domain is,
generally, in accordance with the definitions of Sequences of Proteins of
Immunological
Interest, Kabat, et al.; National Institutes of Health, Bethesda, Md.; 5th
ed.; NIH Publ. No.
91-3242 (1991); Kabat (1978) Adv. Prot. Chem. 32:1-75; Kabat, et al., (1977)
J. Biol. Chem.
252:6609-6616; Chothia et al., (1987) J Mol. Biol. 196:901-917 or Chothia et
al., (1989)
Nature 342:878-883.
[073] As used herein, the term "hypervariable region" refers to the amino acid
residues of
an antibody that are responsible for antigen-binding. The hypervariable region
comprises
amino acid residues from a "complementarity determining region" or "CDR" (i.e.
LCDR1,
LCDR2 and LCDR3 in the light chain variable domain and HCDR1, HCDR2 and HCDR3
in
the heavy chain variable domain). See Kabat et al. (1991) Sequences of
Proteins of
Immunological Interest, 5th Ed. Public Health Service, National Institutes of
Health,
Bethesda, Md. (defming the CDR regions of an antibody by sequence); see also
Chothia and
16

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Lesk (1987) J. Mol. Biol. 196: 901-917 (defming the CDR regions of an antibody
by
structure). As used herein, the term "framework" or "FR" residues refers to
those variable
domain residues other than the hypervariable region residues defined herein as
CDR residues.
[074] As used herein, unless otherwise indicated, "antibody fragment" or
"antigen-binding
fragment" refers to antigen-binding fragments of antibodies, i.e., antibody
fragments that
retain the ability to bind specifically to the antigen bound by the full-
length antibody, e.g.
fragments that retain one or more CDR regions. Examples of antibody binding
fragments
include, but are not limited to, Fab, Fab', F(abt)2, and Fv fragments;
diabodies; linear
antibodies; single-chain antibody molecules, e.g., sc-Fv; nanobodies and multi-
specific
antibodies formed from antibody fragments.
[075] An antibody that "specifically binds to" a specified target protein is
an antibody that
exhibits preferential binding to that target as compared to other proteins,
but this specificity
does not require absolute binding specificity. An antibody is considered
"specific" for its
intended target if its binding is determinative of the presence of the target
protein in a sample,
e.g., without producing undesired results such as false positives. Antibodies,
or binding
fragments thereof, useful in the present invention will bind to the target
protein with an
affinity that is at least two fold greater, preferably at least ten times
greater, more preferably
at least 20 times greater, and most preferably at least 100 times greater than
the affinity with
non-target proteins. As used herein, an antibody is said to bind specifically
to a polypeptide
comprising a given amino acid sequence, e.g. the amino acid sequence of a
mature human
PD-1 or human PD-Ll molecule, if it binds to polypeptides comprising that
sequence but
does not bind to proteins lacking that sequence.
[076] "Chimeric antibody" refers to an antibody in which a portion of the
heavy and/or light
chain is identical with or homologous to corresponding sequences in an
antibody derived
from a particular species (e.g., human) or belonging to a particular antibody
class or subclass,
while the remainder of the chain(s) is identical with or homologous to
corresponding
sequences in an antibody derived from another species (e.g., mouse) or
belonging to another
antibody class or subclass, as well as fragments of such antibodies, so long
as they exhibit the
desired biological activity.
[077] "Human antibody" refers to an antibody that comprises human
immunoglobulin
protein sequences only. A human antibody may contain murine carbohydrate
chains if
produced in a mouse, in a mouse cell, or in a hybridoma derived from a mouse
cell.
17

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Similarly, "mouse antibody" or "rat antibody" refer to an antibody that
comprises only mouse
or rat immunoglobulin sequences, respectively.
[078] "Humanized antibody" refers to forms of antibodies that contain
sequences from non-
human (e.g., murine) antibodies as well as human antibodies. Such antibodies
contain
minimal sequence derived from non-human immunoglobulin. In general, the
humanized
antibody will comprise substantially all of at least one, and typically two,
variable domains,
in which all or substantially all of the hypervariable loops correspond to
those of a non-
human immunoglobulin and all or substantially all of the FR regions are those
of a human
immunoglobulin sequence. The humanized antibody optionally also will comprise
at least a
portion of an immunoglobulin constant region (Fc), typically that of a human
immunoglobulin. The prefix "hum," "hu" or "h" is added to antibody clone
designations
when necessary to distinguish humanized antibodies from parental rodent
antibodies. The
humanized forms of rodent antibodies will generally comprise the same CDR
sequences of
the parental rodent antibodies, although certain amino acid substitutions may
be included to
increase affinity, increase stability of the humanized antibody, or for other
reasons.
[079] "Biotherapeutic agent" means a biological molecule, such as an antibody,
that blocks
ligand/receptor signaling in any biological pathway that supports tumor
maintenance and/or
growth or suppresses the anti-tumor immune response and/or recruits CD8+ T-
cell infiltration
to the tumor.
[080] The terms "cancer," "cancerous," or "malignant" refer to or describe the
physiological
condition in mammals that is typically characterized by unregulated cell
growth. Examples
of cancers associated with one or more of a CD8+ T-cell infiltration density
of fewer than
about 1500, about 1400, about 1300, about 1200, about 1100, about 1000, about
900, about
800, about 700, about 600, or about 500 cells per 1 mm2 sample (or per 1 mL
sample), a low
or negative PD-Li status, and/or a low level of IFNy gene signature
expression, include but
are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma.
[081] Examples of particular cancers that are associated with one or more of a
CD8+ T-cell
infiltration density of fewer than about 1500, about 1400, about 1300, about
1200, about
1100, about 1000, about 900, about 800, about 700, about 600, or about 500
cells per 1 mm2
sample (or per 1 mL sample), a low or negative PD-L1 status, and/or a low
level of IFNy
gene signature expression, include, but are not limited to, melanoma (e.g.,
cutaneous,
metastatic, uveal), non-small cell lung cancer, head and neck cancer (e.g.,
recurrent or
metastatic squamous cell carcinoma of the head and neck), colorectal cancer,
breast cancer
18

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
(e.g., HER2+, HER2- (HR+), triple-negative), ovarian cancer, bladder cancer
(e.g.,
transitional cell cancer, urothelial cancer), prostate cancer (e.g.,
castration-resistant), sarcoma
(e.g., soft tissue, bone), renal cell cancer, gastric cancer, esophageal
cancer, anal canal cancer,
biliary tract cancer or pancreatic cancer.
[082] Examples of particular cancers that are responsive to mono therapy with
a checkpoint
inbibitor (e.g., with anti-PD-Li therapy or anti-PD-1 therapy (e.g.,
pembrolizumab, a
pembrolizumab variant or an antigen-binding fragment thereof)), which may or
may not be
associated with one or more of a CD8+ T-cell infiltration density of fewer
than about 1500,
about 1400, about 1300, about 1200, about 1100, about 1000, about 900, about
800, about
700, about 600, or about 500 cells per 1 mm2 sample (or per 1 mL sample), a
low or negative
PD-Li status, and/or a negative IFNy gene signature, include, but are not
limited to,
melanoma (e.g., cutaneous, metastatic, uveal), lung cancer (e.g., non-small
cell lung cancer,
small cell lung cancer), head and neck cancer (e.g., recurrent or metastatic
squamous cell
carcinoma of the head and neck, nasopharyngeal cancer, thyroid cancer,
salivary cancer,
esophageal cancer), breast cancer (e.g., ER+/HER2- breast cancer, triple-
negative breast
cancer), ovarian cancer, cervical cancer, bladder cancer (e.g., urothelial
cancer), renal cell
cancer, gastrointestinal cancer (e.g., hepatocellular cancer, colorectal
cancer, anal cancer),
biliary tract cancer, multiple myeloma, lymphoma (e.g., mediastinal large B-
cell lymphoma,
Hodgkin's lymphoma) or mesothelioma. In certain embodiments, a particular
cancer type
may have one or more subsets that are hot (i.e., that are not associated with
one or more of a
CD8+ T-cell infiltration density of fewer than about 1500, about 1400, about
1300, about
1200, about 1100, about 1000, about 900, about 800, about 700, about 600, or
about 500 cells
per 1 1111112 sample (or per 1 inL sample), a low or negative PD-Li status,
and/or a negative
IFNy gene signature) and one or more subsets that are cold (i.e., are
associated with one or
more of a CD8+ T-cell infiltration density of fewer than 1000 cells per 1 mm2
sample (or per
1 mL sample), a low or negative PD-L1 status, and/or a negative IFNy gene
signature.
[083] Particularly preferred cancers that may be treated in accordance with
the present
invention include those characterized by one or any combination of: a low CD8+
T-cell
density; a low or negative interferon gamma signature; and a low or negative
PD-Li status.
In certain embodiments, the cancer is "cold," which refers to a cancer
exhibiting a low level
of immune infiltration, e.g., by CD8+ T cells that is typically not
susceptible to anti-PD-1
blockage therapy by converting such tumors to "hot" tumors. In certain
embodiments, a cold
tumor has a CD8+ T-cell density less than or equal to about 3000, e.g., fewer
than about
19

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
3000, about 2900, about 2800, about 2700, about 2600, about 2500, about 2400,
about 2300,
about 22(X), about 2100, about 2000, about 1900, about 1800, about 1700, about
1600,
about1500, about 1400, about 1300, about 1200, about 1100, about 1000, about
900, about
800, about 700, about 600, or about 500 cells per 1 mm2 or 1 mL (i.e., 1 cm3)
sample.
[084] As used herein, a "hot" tumor is a tumor that exhibits higher levels of
immune
infiltration by, e.g., CD8+ T cells, than cold tumors. In certain embodiments,
a hot tumor has
a CD8+ T-cell density of greater than about 3000 cells, e.g., greater than
about 4000 cells or
greater than about 5000 cells per 1 mm2 sample.
[085] CD8+ (cytotoxic) T-cells are generated in the thymus and express the T-
cell receptor.
CD8+ T-cells express a dimeric co-receptor, CD8, usually composed of one CD8a
and one
CD8( 3 chain. CD8+ T-cells recognize peptides presented by MHC Class I
molecules, found
on all nucleated cells. The CD8 heterodimer binds to a conserved portion (the
a3 region) of
MHC Class I during T-celllantigen presenting cell interactions. When a CD8+ T-
cell
recognizes an antigen and becomes activated, it can secrete cytokines, produce
and release
cytotoxic granules, and activate the caspase cascade to kill malignant cells.
[086] A high density of tumor-infiltrating CD8+ T-cells is frequently
associated in the art
with favorable clinical outcomes in a remarkably large spectrum of cancers,
and indicate an
ongoing host immune response, and the prognostic value of a high density of
tumor
infiltrating lymphocytes on clinical outcome has been assessed in a variety of
cancer entities
(Zhang et al (2003) NEjM 348:203-13; Galon et al. (2006) Science 313:1960-64;
Gao et al.
(2007) J Clin Oncol. 25:2586-93; Gooden et al. (2011) Br J Cancer 105:93-103).
[087] The terms "CD8 density," "CD8+ density" or "CD8+ T-cell density" refer
to the
number of CD8+ T-cells present in a sample, e.g., in a tumor sample. In
exemplary
embodiments, a CD8+ T-cell density is the number of cells present in a sample,
e.g., a 1 mm2
sample (e.g., a punch biopsy) or a 1 mL (i.e., 1 cm3) sample (e.g., a liquid
biopsy) of a tumor
from a subject. In certain exemplary embodiments, a low CD8+ T-cell density
(which is
associated with a "cold" tumor) is less than about 3000 cells per 1 mm2 or per
1 mL sample,
less than about 2900 cells per 1 mm2 or per 1 mL sample, less than about 2800
cells per 1
mm2 or per 1 mL sample, less than about 2700 cells per 1 min- or per 1 mL
sample, less than
about 2600 cells per 1 mm2 or per 1 mL sample, less than about 2500 cells per
1 mm2 or per 1
mL sample, less than about 2400 cells per I. mm2 or per I. mL sample, less
than about 2300
cells per 1 nun2 or per 1 mL sample, less than about 2200 cells per 1 mm2 or
per 1 mL
sample, less than about 2100 cells per 1 mm2 or per 1 mL sample, less than
about 2000 cells

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
per 1 mm2 sample, less than about 1900 cells per 1 mm2 sample. less than about
1800 cells
per 1 mm2 or per 1 mL sample. less than about 1700 cells per 1 mm2 or per 1 mL
sample, less
than about 1600 cells per 1 mm2 or per 1 mL sample, less than about 1500 cells
per 1 mm2 or
per 1 mL sample, less than about 1400 cells per 1 mm2 or per 1 mL sample, less
than about
1300 cells per 1 mm2 or per 1 mL sample, less than about 1200 cells per 1 mm2
or per 1 mL
sample, less than about 1100 cells per 1 mm2 or per 1 mL sample, less than
about 1000 cells
per 1 mm2 or per 1 mL sample, less than about 900 cells per 1 mm2 or per 1 mL
sample, less
than about 800 cells per 1 mm2 or per 1 mL sample, less than about 700 cells
per 1 mm2 or
per 1 mL sample, less than about 600 cells per 1 mm2 or per 1 mL sample, less
than about
500 cells per 1 mm2 or per 1 mL sample, less than about 400 cells per 1 mm2 or
per 1 mL
sample, less than about 300 cells per 1 mm2 or per 1 mL sample, less than
about 200 cells per
1 mm2 or per 1 mL sample, or less than about 100 cells per 1 mm2 or per 1 mL
sample. In
certain exemplary embodiments, a low CD8+ T-cell density is between about 3000
and 500
cells per 1 mm2 or per 1 mL sample, between about 2900 and 500 cells per 1 mm2
or per 1
mL sample, between about 2800 and 500 cells per 1 mm2 or per 1 mL sample,
between about
2700 and 500 cells per 1 mm2 or per 1 mL sample, between about 2600 and 500
cells per 1
mm2 or per 1 mL sample, between about 2500 and 500 cells per 1 mm2 or per 1 mL
sample,
between about 2400 and 500 cells per 1 mm2 or per 1 mL sample, between about
2300 and
500 cells per 1 mm2 or per 1 mL sample, between about 2200 and 500 cells per 1
mm2 or per
1 mL sample, between about 2100 and 500 cells per 1 mm2 or per 1 mL sample,
between
about 2000 and 500 cells per 1 mm2 or per 1 mL sample, between about 1900 and
500 cells
per 1 mm2 or per 1 mL sample, between about 1800 and 500 cells per 1 mn12 or
per 1 mL
sample, between about 1700 and 500 cells per 1 mm2 or per 1 mL sample, between
about
1600 and 500 cells per 1 mm2 or per 1 mL sample, 1500 and 500 cells per 1 mm2
or per 1 mL
sample, between about 1400 and 600 cells per 1 mm2 or per 1 mL sample, between
about
1300 and 700 cells per 1 mm2 or per 1 mL sample, between about 1200 and 800
cells per 1
mm2 or per 1 mL sample, between about 1100 and 900 cells per 1 mm2 or per 1 mL
sample,
or between about 1050 and 950 cells per 1 mm2 or per 1 mL sample. In certain
exemplary
embodiments, a low CD8+ T-cell density is between about 10 and 1000 cells per
1 mm2 or
per 1 mL sample, between about 20 and 900 cells per 1 mm2 or per 1 mL sample,
between
about 30 and 800 cells per 1 mm2 or per 1 mL sample, between about 40 and 700
cells per 1
mm2 or per 1 mL sample. between about 50 and 600 cells per 1 mm2 or per 1 mL
sample,
between about 60 and 500 cells per 1 mm2 or per 1 mL sample, between about 70
and 400
21

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
cells per 1 mm2 or per 1 mL sample, between about 80 and 300 cells per 1 mm2
or per 1 mL
sample, or between about 90 and 100 cells per 1 mm2 or per 1 mL sample. In
certain
exemplary embodiments, a sample contains no detectable CD8+ T-cells.
[088] "CDR" or "CDRs" as used herein means complementarity determining
region(s) in an
immunoglobulin variable region, defined using the Kabat numbering system,
unless
otherwise indicated.
[089] A "checkpoint inhibitor" refers to a molecule that totally or partially
reduces, inhibits,
interferes with or modulates one or more checkpoint proteins. Checkpoint
proteins regulate
T-cell activation or function. Numerous checkpoint proteins are known, such as
CTLA-4 and
its ligands CD80 and CD86; and PD1 and its ligands PDL1 and PDL2 (Pardon
(2012) Nature
Reviews Cancer 12: 252-264). These proteins are responsible for co-stimulatory
or inhibitory
interactions of T-cell responses. Checkpoint proteins regulate and maintain
self-tolerance
and the duration and amplitude of physiological immune responses. In certain
embodiments,
checkpoint inhibitors include antibodies or can be derived from antibodies.
[090] "Chemotherapeutic agent" is a chemical compound useful in the treatment
of cancer.
Classes of chemotherapeutic agents include, but are not limited to: alkylating
agents,
antimetabolites, kinase inhibitors, spindle poison plant alkaloids,
cytotoxic/antitumor
antibiotics, topoisomerase inhibitors, photosensitizers, anti-estrogens and
selective estrogen
receptor modulators (SERMs), anti-progesterones, estrogen receptor down-
regulators
(ERDs), estrogen receptor antagonists, leutinizing hormone-releasing hormone
agonists, anti-
androgens, aromatase inhibitors, EGFR inhibitors, VEGF inhibitors, anti-sense
oligonucleotides that that inhibit expression of genes implicated in abnormal
cell proliferation
or tumor growth. Chemotherapeutic agents useful in the treatment methods of
the present
invention include cytostatic and/or cytotoxic agents.
[091] "Clothia" as used herein means an antibody numbering system described in
Al-
Lazikani et al., JMB 273:927-948 (1997).
[092] "Conservatively modified variants" or "conservative substitution" refers
to
substitutions of amino acids in a protein with other amino acids having
similar characteristics
(e.g. charge, side-chain size, hydrophobicity/hydrophilicity, backbone
conformation and
rigidity, etc.), such that the changes can frequently be made without altering
(or substantially
altering) the biological activity or other desired property of the protein,
such as antigen
affinity and/or specificity. Those of skill in this art recognize that, in
general, single amino
acid substitutions in non-essential regions of a polypeptide do not
substantially alter
22

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
biological activity (see, e.g., Watson et al. (1987) Molecular Biology of the
Gene, The
Benjamin/Cummings Pub. Co., p. 224 (4th Ed.)). In addition, substitutions of
structurally or
functionally similar amino acids are less likely to disrupt biological
activity.
[093] "Comprising" or variations such as "comprise," "comprises" or "comprised
of" are
used throughout the specification and claims in an inclusive sense, i.e., to
specify the
presence of the stated features but not to preclude the presence or addition
of further features
that may materially enhance the operation or utility of any of the embodiments
of the
invention, unless the context requires otherwise due to express language or
necessary
implication.
[094] "Consists essentially of," and variations such as "consist essentially
of' or "consisting
essentially of," as used throughout the specification and claims, indicate the
inclusion of any
recited elements or group of elements, and the optional inclusion of other
elements, of similar
or different nature than the recited elements, that do not materially change
the basic or novel
properties of the specified dosage regimen, method, or composition. As a non-
limiting
example, if a gene signature score is defined as the composite RNA expression
score for a set
of genes that consists of a specified list of genes, the skilled artisan will
understand that this
gene signature score could include the RNA expression level determined for one
or more
additional genes, preferably no more than three additional genes, if such
inclusion does not
materially affect the predictive power.
[095] "Framework region" or "FR" as used herein means the immunoglobulin
variable
regions excluding the CDR regions.
[096] "Homology" refers to sequence similarity between two polypeptide
sequences when
they are optimally aligned. When a position in both of the two compared
sequences is
occupied by the same amino acid monomer subunit, e.g., if a position in a
light chain CDR of
two different Abs is occupied by alanine, then the two Abs are homologous at
that position.
The percent of homology is the number of homologous positions shared by the
two sequences
divided by the total number of positions compared x100. For example, if 8 of
10 of the
positions in two sequences are matched or homologous when the sequences are
optimally
aligned then the two sequences are 80% homologous. Generally, the comparison
is made
when two sequences are aligned to give maximum percent homology. For example,
the
comparison can be performed by a BLAST algorithm wherein the parameters of the
algorithm are selected to give the largest match between the respective
sequences over the
entire length of the respective reference sequences.
23

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[097] The following references relate to BLAST algorithms often used for
sequence
analysis: BLAST ALGORITHMS: Altschul, S. F., et at., (1990) J. Mol. Biol.
215:403-410;
Gish, W., et at., (1993) Nature Genet. 3:266-272; Madden, T. L., et al.,
(1996) Meth.
Enzymol. 266:131-141; Altschul, S. F., et at., (1997) Nucleic Acids Res.
25:3389-3402;
Zhang, J., et at., (1997) Genome Res. 7:649-656; Wootton, J. C., et at.,
(1993) Comput.
Chem. 17:149-163; Hancock, J. M. et at., (1994) Comput. App!. Biosci. 10:67-
70;
ALIGNMENT SCORING SYSTEMS: Dayhoff, M. 0., et at., "A model of evolutionary
change in proteins." in Atlas of Protein Sequence and Structure, (1978) vol.
5, suppl. 3. M. 0.
Dayhoff (ed.), pp. 345-352, Natl. Biomed. Res. Found., Washington, D.C.;
Schwartz, R. M.,
et at., "Matrices for detecting distant relationships." in Atlas of Protein
Sequence and
Structure, (1978) vol. 5, suppl. 3. M. 0. Dayhoff (ed.), pp. 353-358, Natl.
Biomed. Res.
Found., Washington, D.C.; Altschul, S. F., (1991) J. Mol. Biol. 219:555-565;
States, D. J., et
at., (1991) Methods 3:66-70; Henikoff, S., et al., (1992) Proc. Natl. Acad.
Sci. USA
89:10915-10919; Altschul, S. F., et at., (1993) J. Mol. Evol. 36:290-300;
ALIGNMENT
STATISTICS: Karlin, S., et at.. (1990) Proc. Natl. Acad. Sci. USA 87:2264-
2268; Karlin, S.,
et at., (1993) Proc. Natl. Acad. Sci. USA 90:5873-5877; Dembo, A., et al.,
(1994) Ann. Prob.
22:2022-2039; and Altschul, S. F. "Evaluating the statistical significance of
multiple distinct
local alignments." in Theoretical and Computational Methods in Genome Research
(S. Suhai,
ed.), (1997) pp. 1-14, Plenum, N.Y.
[098] "Isolated antibody" and "isolated antibody fragment" refers to the
purification status
and in such context means the named molecule is substantially free of other
biological
molecules such as nucleic acids, proteins, lipids, carbohydrates, or other
material such as
cellular debris and growth media. Generally, the term "isolated" is not
intended to refer to a
complete absence of such material or to an absence of water, buffers, or
salts, unless they are
present in amounts that substantially interfere with experimental or
therapeutic use of the
binding compound as described herein.
[099] "Kahat" as used herein means an immunoglobulin alignment and numbering
system
pioneered by Elvin A. Kabat ((1991) Sequences of Proteins of Immunological
Interest, 5th
Ed. Public Health Service, National Institutes of Health, Bethesda, Md.).
[0100] "Monoclonal antibody" or "mAb" or "Mab," as used herein, refers to a
population of
substantially homogeneous antibodies, i.e., the antibody molecules comprising
the population
are identical in amino acid sequence except for possible naturally occurring
mutations that
may be present in minor amounts. In contrast, conventional (polyclonal)
antibody
24

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
preparations typically include a multitude of different antibodies having
different amino acid
sequences in their variable domains, particularly their CDRs, which are often
specific for
different epitopes. The modifier "monoclonal" indicates the character of the
antibody as
being obtained from a substantially homogeneous population of antibodies, and
is not to be
construed as requiring production of the antibody by any particular method.
For example, the
monoclonal antibodies to be used in accordance with the present invention may
be made by
the hybridoma method first described by Kohler et al. (1975) Nature 256: 495,
or may be
made by recombinant DNA methods (see, e.g., U.S. Pat. No. 4,816,567). The
"monoclonal
antibodies" may also be isolated from phage antibody libraries using the
techniques described
in Clackson et al. (1991) Nature 352: 624-628 and Marks et al. (1991) J. Mol.
Biol. 222: 581-
597, for example. See also Presta (2005) J. Allergy Clin. Immunol. 116:731.
[0101] "Interferon gamma" and "IFNy' (also called immune or type 11
interferon), refers to a
pleiotropic cytokine involved in the regulation of nearly all phases of immune
and
inflammatory responses, including the activation, growth and differentiation
of T-cells, B-
cells, macrophages, NK cells and other cell types such as endothelial cells
and fibroblasts.
IFNy enhances MHC expression on antigen-presenting cells, and also plays an
important role
in activating lymphocytes to enhance anti-tumor effects.
[0102] 1FNy can contribute to the containment of tumor progression and growth
by
increasing tumor antigen presentation to tumor-specific T-cells and increasing
susceptibility
to NK cytotoxicity. In addition to promoting an immune response to the tumor,
IFN-y can
also induce expression of tumor suppressing factors.
[0103] "Genetically modified oncolytic virus," as used herein, refers to an
oncolytic virus
that has been modified as compared to a wild-type version of the virus,
typically to remove
and/or insert one or more genes. A preferred genetically modified oncolytic
virus of the
invention is talimogene laherparepvec, also known as IMLYGIC (INN =
talimogene
laherparepvec), a genetically engineered herpes virus that is commercially
available from
Amgen Inc. (Thousand Oaks, CA). Talimogene laherparepvec is described in,
e.g., WO
2014036412, incorporated herein by reference in its entirety for all purposes.
[0104] Talimogene laherparepvec, HS V-1 (strain JS1) ICP34.5-/ICP47-/hGM-CSF
-
(previously known as OncoVex m i (-F), s an intratumorally delivered
oncolytic
immunotherapy comprising an immune-enhanced HSV-1 that selectively replicates
in solid
tumors. (Lui et al., Gene Therapy, 10:292-303, 2003; US Patent No. 7,223,593
and US
Patent No. 7,537,924.) The HSV-1 was derived from Strain JS1 as deposited at
the European

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
collection of cell cultures (ECAAC) under accession number 01010209. In
talimogene
laherparepvec, the HSV-1 viral genes encoding ICP34.5 have been functionally
deleted.
Functional deletion of ICP34.5, which acts as a virulence factor during HSV
infection, limits
replication in non-dividing cells and renders the virus non-pathogenic. In
addition, in
talhnogene laherparepvec. the HSV-1 viral gene encoding ICP47 (which blocks
viral antigen
presentation to major histocompatibility complex class I and II molecules) has
been
functionally deleted. Functional deletion of TCP47 also leads to earlier
expression of US11, a
gene that promotes virus growth in tumor cells without decreasing tumor
selectivity. Finally,
the coding sequence for human GM-CSF, a cytokine involved in the stimulation
of immune
responses, has been inserted into the viral genome of talimogene
laherparepvec. The
insertion of the gene encoding human GM-CSF is such that it replaces nearly
all of the
ICP34.5 gene, ensuring that any potential recombination event between
talimogene
laherparepvec and wild-type virus could only result in a disabled, non-
pathogenic virus and
could not result in the generation of wild-type virus carrying the gene for
human GM-CSF.
The HSV thymidine kinase (TK) gene remains intact in talimogene laherparepvec,
which
renders the virus sensitive to anti-viral agents such as acyclovir. Therefore,
acyclovir can be
used to block talimogene laherparepvec replication, if necessary.
[0105] In a prior phase 3 clinical trial, intratumoral injection of talimogene
laherparepvec
into melanoma metastases improved the durable response rate compared with
subcutaneous
GM-CSF in patients with advanced melanoma (Andtbacka et al. (2015). Talimogene
Laherparepvec Improves Durable Response Rate in Patients With Advanced
Melanoma. J
Clin Oncol 33, 2780-2788). Promising anti-tumor activity was also demonstrated
when
talimogene laherparepvec was given together with the checkpoint inhibitor
ipilimumab,
which blocks the cytotoxic T-cell associated-antigen 4 (CTLA-4) (Chesney, J.,
Collichio, F.,
Andtbacka, R.H., Puzanov, I.. Glaspy, J.A., Milhem, M., Hamid, 0., Cranmer,
L., Saenger,
Y., Ross, M., et al. (2016). Interim safety and efficacy of a randomized
(1:1), open-label
phase 2 study of talimogene laherparepvec (T) and ipilimumab (I) vs I alone in
unresected,
stage BIB- IV melanoma. Ann Oncol 27 (6), 379-400; Puzanov, I., Milhem, M.M.,
Minor,
D., Hamid, 0., Li, A., Chen, L., Chastain, M., Gorski, K.S., Anderson, A.,
Chou, J.. et al.
(2016). Talimogene Laherparepvec in Combination With Ipilimumab in Previously
Untreated, Unresectable Stage ITTB-IV Melanoma. J Clin Oncol 34, 2619-2626).
[0106] Talimogene laherparepvec (IMLYGIC ) was approved as a monotherapy
treatment
for metastatic melanoma in the U.S., European Union, and Australia in 2015. In
OPTiM, a
26

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
multicenter, phase 3 clinical trial that enrolled patients with metastatic
melanoma that could
not be surgically removed, patients who received talimogene laherparepvec were
significantly more likely to experience a durable response compared with
patients who
received the comparator therapy, GM-CSF. (Andtbacka RHI, et al., J. Clin
Oncol., 33:2780-
2788 (2015)).
[0107] In addition, the safety of ICP34.5-functionally deleted HSVs has been
shown in
multiple clinical studies (MacKie et at, Lancet 357: 525-526, 2001; Marked et
al, Gene Ther
7: 867-874, 2000; Rampling et at, Gene Ther 7:859-866, 2000; Sundaresan et at,
J. Virol 74:
3822-3841, 2000; Hunter et al, J Virol Aug; 73(8): 6319- 6326, 1999).
[0108] Talimogene laherparepvec produces a direct oncolytic effect by
replication of the
virus in the tumor, and induction of an anti-tumor immune response enhanced by
the local
expression of GM-CSF. Intended clinical effects include, but are not limited
to, the
destruction of injected tumors; the destruction of local, local-regional, and
distant uninjected
tumors; a reduction in the development of new metastases; a reduction in the
rate of overall
progression; and prolonged overall survival.
[0109] Talimogene laherparepvec has been tested for efficacy in a variety of
in vitro (cell
line) and in vivo murine tumor models and has been shown to eradicate tumors
or
substantially inhibit their growth at doses comparable to those used in
clinical studies. Non-
clinical evaluation has also confirmed that GM-CSF enhances the immune
response
generated, enhancing both injected and uninjected tumor responses, and that
increased
surface levels of MHC class I molecules result from the deletion of ICP47.
Talimogene
laherparepvec has been injected into normal and tumor-bearing mice to assess
its safety. In
general, the virus has been well tolerated, and doses up to 1 x 108 PFU/dose
have given no
indication of any safety concerns. (See, for example, Liu et al., Gene Ther
10: 292-303,
2003).
[0110] Clinical studies have been or are being conducted in several advanced
tumor types
(advanced solid tumors, melanoma, squamous cell cancer of the head and neck,
and
pancreatic cancer), with over 4(X) subjects treated with talimogene
laherparepvec (see, for
example, Hu et al., Clin Can Res 12: 6737-6747, 2006; Harrington et al., J
Clin Oncol.
27(15a):abstract 6018, 2009; Kaufman et al., Ann Surgic Oncol. 17: 718-730,
2010; Kaufman
and Bines, Future Oncol. 6(6): 941-949, 2010).
27

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0111] "Oligonucleotide" refers to a nucleic acid that is usually between 5
and 100
contiguous bases in length, and most frequently between 10-50, 10-40, 10-30,
10-25, 10-20,
15-50, 15-40, 15-30, 15-25, 15-20, 20-50, 20-40, 20-30 or 20-25 contiguous
bases in length.
[0112] "Patient" or "subject" refers to any single subject for which therapy
is desired or that
is participating in a clinical trial, epidemiological study or used as a
control, including
humans, non-human primates, mammalian veterinary patients such as cattle,
horses, dogs,
cats and the like, and research animals such as non-human primates, rats,
mice, dogs, rabbits
and the like.
[0113] Pembrolizumab is a humanized monoclonal antibody against that binds to
and blocks
PD-1. Pembrolizumab works by increasing the ability of the body's immune
system to help
detect and fight tumor cells by blocking the interaction between PD-1 and its
ligands, PD-Li
and PD-L2, thereby activating T lymphocytes which may affect both tumor cells
and healthy
cells.
[0114] Pembrolizumab monotherapy is known to treat melanoma, non-small cell
lung cancer
and squamous cell carcinoma of the head and neck in affected individuals
having higher
densities of baseline CD8+ T-cell infiltrations, IFNy gene signature and PD-Li
expression
than levels found in non-responsive individuals. This was understood by others
in the art to
limit the utility of pembrolizumab in individuals with a low CD8+ T-cell
density, a negative
IFNy gene signature and/or a low or negative PD-Li status.
[0115] As used herein, "pembrolizumab" refers to a commercially available
monoclonal
antibody under the proprietary name of ICEYTRUDA (Merck Sharp & Dohme Corp.,
Whitehouse Station, NJ), described in W02016196173 and U.S. Pat. Nos.
8,354,509 and
8,900,587, incorporated herein by reference in their entireties for all
purposes, as well as
variants and antigen-binding fragments thereof Pembrolizumab can be
characterized by one
or any combination of the heavy chain domain, light chain domain, heavy chain
variable
domain, light chain variable domain, heavy chain complementarity-determining
and light
chain complementarity-determining sequences described infra.
[0116] Pembrolizumab can comprise a heavy chain sequence set forth as
QVQLVQSGVEVKKPGASVKVSCKASGYTFTNYYMYWVRQAPGQGLEWMGGINPS
NGGTNFNEICFKNRVTLTTDSSTTTAYMELKSLQFDDTAVYYCARRDYRFDMGFDY
WGQGTTVTVSSASTKGPSVFPLAPCSRSTSESTAALGCLVICDYFPEPVTVSWNSGAL
TSGVHTFPA VLQSSGLY SLSS V VTVPS SSLGTKTYTCN VDHKPSNTK VDKRVESKYG
PPCPPCPAPEFLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSQEDPEVQFNWYVDG
28

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
VEVHNAKTKPREEQFNSTYRVVSVLTVLHQDWLNGKEYKC KVSNKG LPSSIEKTIS K
AKGQPREPQVYTLPPS QEEMTICNQVSLTCLVKGFYPSDIA VEWESNG QPENNYKTTP
PVLDSDGSFFLYSRLTVDKSRWQEGNVFSCS VMHEALHNHYTQKSLSLSLGK (SEQ
ID NO:1), and a light chain sequence set forth as
EIVLTQSPATLSLSPGERATLSCRASKGVSTSGYSYLHWYQQKPGQ
APRLLIYLASYLESGVPARFSGSGSGTDFTLTISSLEPEDFAVYYCQHSRDLPLTFGGG
TKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWK VDNALQSGN
SQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGEC
(SEQ ID NO:2).
[0117] Pembrolizumab can comprise a heavy chain variable (VH) domain sequence
set forth
as
QVQLVQSGVEVKICPGASVKVSC KASGYTFTNYYMYWVRQAPGQGLEWMGGIN PS
NGGTNFNEICFKNRVTLTTDSSTTTA YMELKSLQFDDTAVY YCARRDYRFDMGFDY
WGQGTTVTVSS (SEQ Ill NO:3), and a light chain variable (VL) domain set forth
as
EIVLTQSPATLSLSPGERATLSCRASKGVSTSGYSYLHWYQQKPGQAPRLLIYLASYL
ESGVPARFSGSGSGTDI-TLTISSLEPEDFAVY YCQHSRDLPLTFGGGTKVEIK (SEQ ID
NO:4).
[0118] Pembrolizumab can comprise the following heavy chain complementarity-
determining regions (HCDRs): NYYMY (HCDR1, SEQ ID NO:5); GINPSNGGTNFN
(HCDR2, SEQ Ill NO:6); and RDYRFDMGFDY (HCDR3, SEQ ID NO:7).
[0119] Pembrolizumab can comprise the following light chain complementarity-
determining
regions (LCDRs): RASKGVSTSGYSYLH (LCDR1, SEQ ID NO:8); LASYLES (LCDR2,
SEQ ID NO:9); and QHSRDLPLT (LCDR3, SEQ ID NO:10).
[0120] In certain embodiments, pembrolizumab, a pembrolizumab variant or an
antigen-
binding fragment thereof is provided comprising heavy chain CDRs SEQ ID NOs:
5, 6 and 7
and light chain CDRs of SEQ ID NOs: 8, 9 and 10.
[0121] In other embodiments, pembrolizumab, a pembrolizumab variant or an
antigen-
binding fragment thereof is provided comprising heavy chain and light chain
CDR sequences
from a VH/VL sequence pair of SEQ ID NO:3 and SEQ ID NO:4.
[0122] In still other preferred embodiments, pembrolizumab, a pembrolizumab
variant or an
antigen-binding fragment thereof is provided comprising a heavy chain variable
region
comprising SEQ ID NO:3 or a variant thereof and/or a light chain variable
region comprising
SEQ ID NO:4 or a variant thereof. In other embodiments, the pembrolizumab
variant or
29

CA 03057157 2019-09-18
WO 2018/201028 PCT/US2018/029915
antigen-binding fragment thereof comprises a heavy chain variable region
comprising as
sequence with at least 80% sequence homology or identity (e.g., 80%, 85%, 90%,
95%, 98%
or 99%) to SEQ Ill NO:3 and/or a light chain variable region comprising a
sequence with at
least 80% sequence homology or identify (e.g., 80%, 85%, 90%, 95%, 98% or 99%)
to SEQ
ID NO:4.
[0123] As used herein, a "variant of a heavy chain variable region sequence"
is a sequence
that is identical to the reference sequence, except having up to 17
conservative amino acid
substitutions in the framework region (i.e., outside of the CDRs), and
preferably having fewer
than ten, nine, eight, seven, six or five conservative amino acid
substitutions in the
framework region. As used herein, a "variant of a light chain variable region
sequence" is a
sequence that is identical to the reference sequence, except having up to five
conservative
amino acid substitutions in the framework region (i.e., outside of the CDRs),
and preferably
having fewer than four, three or two conservative amino acid substitution in
the framework
region.
[0124] In still other embodiments, pembrolizumab, a pembrolizumab variant or
an antigen-
binding fragment thereof is provided comprising a heavy chain comprising SEQ
ID NO:1 or
a variant thereof and/or a light chain comprising SEQ ID NO:2 or a variant
thereof. In other
embodiments, the pembrolizumab variant or antigen-binding fragment thereof
comprises a
heavy chain comprising as sequence with at least 80% sequence homology or
identity (e.g.,
80%, 85%, 90%, 95%, 98% or 99%) to SEQ ID NO:1 and/or a light chain comprising
a
sequence with at least 80% sequence homology or identify (e.g., 80%, 85%, 90%,
95%, 98%
or 99%) to SEQ ID NO:2.
[0125] As used herein, a "pembrolizumab variant" refers to a monoclonal
antibody which
comprises heavy chain and light chain sequences that are identical to those of
pembrolizumab, except for having up to five conservative amino acid
substitutions in the
framework region (i.e., outside of the CDRs), and preferably has less than
four, three or two
conservative amino acid substitution in the framework region, and having up to
17
conservative amino acid substitutions in the framework region (i.e., outside
of the CDRs),
and preferably has less than ten, nine, eight, seven, six or five conservative
amino acid
substitutions in the framework region, and preferably has less than four,
three or two
conservative amino acid substitution in the framework region. In other
words,
pembrolizumab and a pembrolizumab variant comprise identical CDR sequences,
but differ
from each other due to having a conservative amino acid substitution at no
more than three or

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
six other positions in their full length light and heavy chain sequences,
respectively. A
pembrolizumab variant is substantially the same as or better than
pembrolizumab with respect
to the following properties: binding affinity to PD-1 and neutralizing effect
in vivo.
[0126] In certain embodiments, biosimilars of pembrolizumab are provided. As
used herein,
the term "biosimilar" is used in a manner that is consistent with the working
definition
promulgated by the U.S. Food and Drug Administration, which defines a
biosimilar product
to be one that is "highly similar" to a reference product (despite minor
differences in
clinically inactive components). In practice, there can be no clinically
meaningful differences
between the reference product and the biosimilar product in terms of safety,
purity, and
potency (Public Health Service (PHS) Act 262). In certain embodiments, a
double-blind,
single-dose comparative pharmacokinetic (PK) crossover study is performed to
compare
pembrolizumab with a candidate biosimilar antibody to determine comparable
bioavailability.
[0127] As used herein, the term "reference product," is used to refer to
commercially
available pembrolizumab.
[0128] The PD-1 receptor (also known as CD279) is expressed on the surface of
activated T-
eens. Its ligands, PD-Li (B7-H1; CD274) and PD-L2 (B7-DC; CD273), are commonly
expressed on the surface of dendritic cells or macrophages. PDI and PD-Li /PD-
L2 belong to
the family of immune checkpoint proteins that act as co-inhibitory factors,
which can halt or
limit the development of the T cell response. PD1/PD-L1 interaction ensures
that the
immune system is activated only at the appropriate time in order to minimize
the possibility
of chronic autoimmune inflammation. When PD-Li binds to PD-1, an inhibitory
signal is
transmitted into the T-cell, which reduces cytokine production and suppresses
T-cell
proliferation. Tumor cells exploit this immune-checkpoint pathway as a
mechanism to evade
detection and inhibit the immune response.
[0129] PD-L1 is commonly overexpressed on tumor cells or on non-transformed
cells in the
tumor microenvironment. PD-Li expressed on the tumor cells binds to PD-1
receptors on the
activated T-cells, which leads to the inhibition of the cytotoxic T-cells.
These deactivated T-
cells remain inhibited in the tumor microenvironment. The PD1/PD-L1 pathway
represents
an adaptive immune resistance mechanism that is exerted by tumor cells in
response to
endogenous anti-tumor activity.
[0130] As used herein, "PD-Li status" or "PD-Li expression status" refers to
the level of
PD-L1 present in a sample, e.g., in a tumor sample. In certain embodiments, PD-
L1 status is
expressed as a "tumor proportion score" or "TPS," which refers to the
percentage of tumor
31

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
cells in a sample that express a detectable level of PD-Li. (See Garon et al.
(2015) NEJM
372: 2018-28.) A PD-Li status is "underexpressed" or "reduced" or "low" if PD-
Li is
expressed in between about 1% and about 49% of tumor cells in a sample, i.e.,
the PD-Ll
status is between about 1% and about 49%. A PD-Li status is "negative" if PD-
L1 is
expressed in fewer than about 1% of tumor cells in a sample, i.e., the PD-L1
status is less
than about 1%. A PD-Li status is "high" if PD-Li is expressed in about 50% or
more of the
tumor cells in a sample, i.e., the PD-Li status is about 50% or greater.
[0131] In some preferred embodiments, an underexpressed, reduced or low PD-L1
status is
less than about 50%, or is about 49%, about 48%, about 47%, about 46%, about
45%, about
44%, about 43%, about 42%, about 41%, about 40%, about 39%, about 38%, about
37%,
about 36%, about 35%, about 34%, about 33%, about 32%, about 31%, about 30%,
about
29%, about 28%, about 27%, about 26%, about 25%, about 24%, about 23%, about
22%,
about 21%, about 20%, about 19%, about 18%, about 17%, about 16%, about 15%,
about
14%, about 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%,
about
6%, about 5%, about 4%, about 3%, about 2%, or about 1%. In particularly
preferred
embodiments, an underexpressed, reduced or low PD-L1 status is about 10%,
about 9%,
about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, or about
1%. In
some preferred embodiments, an underexpressed, reduced or low PD-Li status is
between
about 1% and about 10%, between about 1% and about 9%, between about 1% and
about 8%,
between about 1% and about 7%, between about 1% and about 6%, between about 1%
and
about 5%, between about 1% and about 4%, between about 1% and about 3%,
between about
1% and about 2%, between about 2% and about 6%, between about 3% and about 7%,
between about 4% and about 8%, or between about 5% and about 9%.
[0132] In some preferred embodiments, a negative PD-Li status is less than
about 1%, or is
about 0.9%, about 0.8%, about 0.7%, about 0.6%, about 0.5%, about 0.4%, about
0.3%, about
0.2%, about 0.1%, about 0.05%, about 0.01%, about 0.005%, about 001%, or about
0% (i.e.,
none of the tumor cells have a detectable level of PD-Li).
[0133] In some preferred embodiments, a high PD-Li status is about 50% or
greater, e.g., is
about 55%, about 60%, about 65%, about 70%, about 75%, about 80%, about 85%,
about
90%, about 95%, about 96%, about 97%, about 98%, about 99%, or about 100%
(i.e., all
of the tumor cells have a detectable level of PD-L1).
[0134] In certain preferred embodiments, PD-L1 expression is detected using a
diagnostic
anti-human PD-Li antibody, or antigen-binding fragment thereof, in an IHC
assay on an
32

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
FFPE or frozen tissue section of a tumor sample removed from the subject. In
certain
embodiments, PD-Li expression may be assessed in a sample (e.g., a tumor
sample) using a
PD-Li IHC 22C3 pharmDx immunohistochemistry assay (Dako North America,
Carpinteria,
CA). (See Daud, A.I., Wolchok, J.D., Robert, C., Hwu, W.J., Weber, J.S.,
Ribas, A., Hodi,
F.S., Joshua, A.M., Kefford, R.. Hersey, P., et al. (2016). Programmed Death-
Ligand 1
Expression and Response to the Anti-Programmed Death 1 Antibody Pembrolizumab
in
Melanoma. J Clin Oncol 34, 4102-4109.) Typically, a subject's physician would
order a
diagnostic test to determine PD-L1 expression in a tumor tissue sample removed
from the
patient prior to initiation of treatment with pembrolizumab, pembrolizumab
variant and/or
antigen-binding fragment thereof and/or talimogene laheiparepvec, but it is
envisioned that
the physician could order the first or subsequent diagnostic tests at any time
after initiation of
treatment, such as for example after completion of a treatment cycle.
[0135] "Primary pembrolizumab antibody" and "primary pembrolizumab variant
antibody"
as used herein, refer to an antibody that binds specifically to PD-Li in a
tissue section, and is
generally the first antibody used in an IHC assay of PD-Li expression in a
tumor sample. In
one embodiment, the primary antibody is the only antibody used in the 1HC
assay.
[0136] "Secondary antibody," as used herein, refers to an antibody that binds
specifically to a
primary pembrolizumab antibody or a primary pembrolizumab variant antibody,
thereby
forming a bridge between the primary antibody and a subsequent detection
reagent, if any.
The secondary antibody is generally the second antibody used in an 1HC assay
of PD-Li
expression in a tumor sample.
[0137] "Patient," as used herein, refers to a subject having a cancer that is
treated by a
combination of pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment
thereof and talimogene lahetparepvec.
[0138] In certain embodiments, a patient has a cancer that is associated with
one or more of a
CD8+ T-cell infiltration density of fewer than about 1500, about 1400, about
1300, about
1200, about 1100, about 1000, about 900, about 800, about 700, about 600, or
about 500 cells
per 1 mm2 sample (or per 1 mL sample), a low or negative PD-Li status, and a
low level of
IFNy gene signature, including, but not limited to, melanoma (e.g., cutaneous,
metastatic,
uveal), non-small cell lung cancer, head and neck cancer (e.g., recurrent or
metastatic
squamous cell carcinoma of the head and neck), colorectal cancer, breast
cancer (e.g.,
HER2+, HER2- (HR+), triple-negative), ovarian cancer, bladder cancer (e.g.,
transitional cell
cancer, urothelial cancer), prostate cancer (e.g., castration-resistant),
sarcoma (e.g., soft
33

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
tissue, bone), renal cell cancer, gastric cancer, esophageal cancer, anal
canal cancer, biliary
tract cancer or pancreatic cancer.
[0139] in other embodiments, a patient has a cancer that is responsive to
monotherapy with a
checkpoint inbibitor (e.g., with anti-PD-L1 therapy or anti-PD-1 therapy
(e.g.,
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)), which
may optionally be associated with one or more of a CD8+ T-cell infiltration
density of fewer
than about 1500, about 1400, about 1300, about 1200, about 1100, about 1000,
about 900,
about 800, about 700, about 600, or about 500 cells per 1 mm2 sample (or per 1
rnL sample),
a low or negative PD-Li status, and a low level of IFNy gene signature,
including, but not
limited to, melanoma (e.g., cutaneous, metastatic, uveal), lung cancer (e.g.,
non-small cell
lung cancer, small cell lung cancer), head and neck cancer (e.g., recurrent or
metastatic
squamous cell carcinoma of the head and neck, nasopharyngeal cancer, thyroid
cancer,
salivary cancer, esophageal cancer), breast cancer (e.g., ER+/HER2- breast
cancer, triple-
negative breast cancer), ovarian cancer, cervical cancer, bladder cancer
(e.g., urothelial
cancer), renal cell cancer, gastrointestinal cancer (e.g., hepatocellular
cancer, colorectal
cancer, anal cancer), biliary tract cancer, multiple myeloma, lymphoma (e.g.,
mediastinal
large B-cell lymphoma, Hodgkin's lymphoma) or mesothelioma.
[0140] "Probe" as used herein means an oligonucleotide that is capable of
specifically
hybridizing under stringent hybridization conditions to a transcript expressed
by a gene of
interest, and in some preferred embodiments, specifically hybridizes under
stringent
hybridization conditions to the particular transcript for the gene of
interest.
[0141] `RECIST 1.1 Response Criteria" as used herein means the definitions set
forth in
Eisenhauer et al., E. A. et al., Eur. J Cancer 45:228-247 (2009) for target
lesions or non-target
lesions, as appropriate, based on the context in which response is being
measured.
[0142] "Reference IFN-y gene signature score," as used herein, means the score
for an IFN-y
gene signature that has been determined to divide at least the majority of
responders from at
least the majority of non-responders in a reference population of subjects who
have the same
tumor type as a test subject and who has been treated with a combination
therapy of
pembrolizumab, a pembrolizumab variant and/or an antigen-binding fragment
thereof and
talimogene laherparepvec. Preferably, at least any of 60%, 70%, 80%, or 90% of
responders
in the reference population will have an IFN-y gene signature score that is
below the selected
reference score (e.g., a pre-specified threshold of a control panel) (i.e., a
negative IFNy gene
signature), while the IFN-y gene signature for at least any of 60%, 70% 80%,
90% or 95% of
34

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
the non-responders in the reference population will be greater than the
selected reference
score (i.e., a positive IFNy gene signature). In some preferred embodiments,
responders in
the reference population are defined as subjects who achieved a partial
response (PR) or
complete response (CR) as measured by RECIST 1.1 criteria, and non-responders
are defined
as not achieving any RECIST 1.1 clinical response.
[0143] "Responder patient" when referring to a specific anti-tumor response to
treatment
with a combination therapy described herein, means the patient exhibited the
anti-tumor
response.
[0144] "Sample" when referring to a tumor or any other biological material
referenced
herein, means a sample that has been removed from the subject.
[0145] "Sustained response" means a sustained therapeutic effect after
cessation of treatment
with a therapeutic agent, or a combination therapy described herein. In some
embodiments,
the sustained response has a duration that is at least the same as the
treatment duration, or at
least 1.5, 2.0, 2.5 or 3 times longer than the treatment duration.
[0146] "Tissue Section" refers to a single part or piece of a tissue sample,
e.g., a thin slice of
tissue cut from a sample of a normal tissue or of a tumor.
[0147] "Treat" or "treating" a cancer as used herein means to administer
pembrolizumab, a
pembrolizumab variant or an antigen-binding fragment thereof and talimogene
laherparepvec,
and/or another therapeutic agent, to a subject having a cancer, or diagnosed
with a cancer, to
achieve at least one positive therapeutic effect, such as for example, reduced
number of
cancer cells, reduced tumor size, reduced rate of cancer cell infiltration
into peripheral
organs, or reduced rate of tumor metastasis or tumor growth. Positive
therapeutic effects in
cancer can be measured in a number of ways (See, W. A. Weber, J. Null. Med.
50:1S-10S
(2009); Eisenhauer et al., supra). In some preferred embodiments, response
to
pembrolizumab, a pembrolizumab variant and/or an antigen-binding fragment
thereof, and/or
talimogene laherparepvec is assessed using RECIST 1.1 criteria. In some
embodiments, the
treatment achieved by a therapeutically effective amount is any of a partial
response (PR), a
complete response (CR), progression free survival (PFS), disease free survival
(DFS),
objective response (OR) or overall survival (OS). In some preferred
embodiments, a CD8+
cell density, a gene signature biomarker and/or a low or negative PD-L1 status
of the
invention predicts whether a subject with a solid tumor is likely to achieve a
PR or a CR. The
dosage regimen of a therapy described herein that is effective to treat a
cancer patient may
vary according to factors such as the disease state, age, and weight of the
patient, and the

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
ability of the therapy to elicit an anti-cancer response in the subject. While
an embodiment of
the treatment method, medicaments and uses of the present invention may not be
effective in
achieving a positive therapeutic effect in every subject, it should do so in a
statistically
significant number of subjects as determined by any statistical test known in
the art such as
the Student's t-test, the chi2-test, the U-test according to Mann and Whitney,
the ICruskal-
Wallis test (H-test), Jonckheere-Terpstra-test and the Wilcoxon-test.
[0148] "Tumor" as it applies to a subject diagnosed with, or suspected of
having a cancer,
refers to a malignant or potentially malignant neoplasm or tissue mass of any
size, and
includes primary tumors and secondary neoplasms. A solid tumor is an abnormal
growth or
mass of tissue that usually does not contain cysts or liquid areas. Different
types of solid
tumors are named for the type of cells that form them. Examples of solid
tumors are
sarcomas, carcinomas, and lymphomas. Leukemias (cancers of the blood)
generally do not
form solid tumors (National Cancer Institute, Dictionary of Cancer Terms).
[0149] "Tumor burden" also referred to as "tumor load," refers to the total
amount of tumor
material distributed throughout the body. Tumor burden refers to the total
number of cancer
cells or the total size of tumor(s) throughout the body, including lymph nodes
and bone
narrow. Tumor burden can be determined by a variety of methods known in the
art, such as,
e.g., by measuring the dimensions of tumor(s) upon removal from the subject,
e.g., using
calipers, or while in the body using imaging techniques, e.g., ultrasound,
bone scan,
computed tomography (CT) or magnetic resonance imaging (MRI) scans.
[0150] The term "tumor size" refers to the total size of the tumor which can
be measured as
the length and width of a tumor. Tumor size may be determined by a variety of
methods
known in the art, such as, e.g. by measuring the dimensions of tumor(s) upon
removal from
the subject, e.g., using calipers, or while in the body using imaging
techniques, e.g., bone
scan, ultrasound, CT or MRI scans.
[0151] "Variable regions" or "V region" as used herein means the segment of
IgG chains
which is variable in sequence between different antibodies. It extends to
Kabat residue 109 in
the light chain and 113 in the heavy chain.
[0152] In some embodiments of the treatment methods, medicaments and uses of
the
invention disclosed herein, the individual is a human and the cancer
correlates with a low or
negative PD-Li status and/or a low or negative CD8+ T-cell density, including,
but not
limited to: melanoma (generally highly T-cell infiltrated high% PD-L1+; subset
PD-LI/TIL
low, Tumeh et al Nature 2014; Daud, JCO 2016); non-small cell lung cancer
(some high%
36

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
PD-L1+; subset PD-Ll/TIL low, Topalian NEJM 2012; Garon et al NEJM 2015;
Ameratunga
PlosOne 2016); recurrent or metastatic squamous cell carcinoma of the head and
neck; HPV
positive head and neck cancer or HPV negative head and neck cancer (generally
highly T-cell
infiltrated high% PD-L1+; subset PD-L1/TIL low apparently true for both HPV+
and HPV-
head and neck cancers, Lyford-Pyke CanRes 2013; Ma1m Head Neck 2015; Mandal et
al JCI
2016); colorectal cancer (mismatch repair deficient CRC/ Lynch syndrome highly
PD-L1+
TIL infiltrated; MSI stable CRC generally cold tumors, Le et. at NEJM 2015;
Maby CanRes
2015); HER2+ breast cancer (CD8+ high (61%) Foxp3 Treg high; lymphocyte
predominant
breast cancers (16%), Stanton JAMA (2016)); HER2- (BR+) breast cancer (CD8+
mid(43%)
lower Foxp3 Treg; lymphocyte predominant breast cancers (6%), Stanton JAMA
(2016));
triple-negative breast cancer (CD8+ high (60%) Foxp3 Treg high; lymphocyte
predominant
breast cancers (20%), Stanton JAMA (2016)); ovarian cancer (prognostic data
around TIL
similar to melanoma; response rate 15-40% in Hamanishi jCO3 Hamanishi JCO,
2015;
Mandai IJCO 2016); bladder cancer (correlation with PD-L 1 ; possible TIL
bladder cancer
correlation for checkpoint inhibitors, Powles Nature 2014 (Atezo); Kim ICU
2016); uveal
melanoma (bmx data presented Piperno-Neumann ASCO 2016, uveal melanoma);
castration
resistant prostate cancer (Paucity of PD-Li expression in prostate cancer:
innate and adaptive
immune resistance, Martin Prostate Cancer and Prostatic Diseases 2015 Kim ICU
2016); soft
tissue or bone sarcoma (modest response rate; prognostic PD-Li / TIL data
available ore
digging needed to find clinical data with TIL vs checkpoint inhibitor response
-, L. Paoluzzi
Clinical Sarcoma Research 2016; D'Angelo SP Human Pathol 2015); and renal cell
cancer
(pembrolizumab primarily used in combination in RCC: Ipi, axitinib, anti-VEGF
etc.
generally highly T-cell infiltrated high% PD-L1+; subset PD-Ll/TIL low, Taube
Clin Can
Res 2014).
[0153] In other embodiments of the treatment methods, medicaments and uses of
the
invention disclosed herein, the individual is a human and the cancer
correlates with a low or
negative IFNy mRNA signature, including, but not limited to: melanoma (ORR/PFS
correlation with 1FNy signature, Ayers et at JITC 2015); head and neck cancer
("Inflamed
phenotype" signatures are strong predictors of clinical benefit from anti-PD-1
treatment for
HNSCC even among a group of patients already considered to be PD-L1+; Clinical
trial
information: NCT01848834, Seiwert ASCO et at JITC); gastric cancer (PFS
correlation with
IFNy signature, Ayers et at JITC 2015); esophageal cancer (Overall, those with
higher
signature scores experienced a more robust response to pembrolizumab and
substantial delays
37

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
in progression; in the non-inflamed, low score group, the ORR was 11% compared
with an
43% for those with a higher signature score, Doi et al GCS 2016; this RNA
profiling data set
from 5 cancers adds to the growing body of evidence that tumor infiltration
with activated T-
cells is a prerequisite for response to PD-1 checkpoint blockade, and
demonstrates that T-cell
inflamed gene expression signatures are pan-cancer predictors of clinical
benefit from anti-
PD-1 treatment, Piha-Paul et al. ASCO 2016); anal canal cancer (as per Piha-
Paul et al.
ASCO 2016); biliary tract cancer (as per Piha-Paul et al. ASCO 2016);
colorectal cancer (as
per Piha-Paul et al. ASCO 2016); ovarian cancer (as per Piha-Paul et al. ASCO
2016); and
transitional cell cancer (similar to melanoma, SCCHN, and gastric with regard
to predictive
value of IFN7 signature).
[0154] In other embodiments of the above treatment method, medicaments and
uses, the
individual is a human, and the cancer correlates with low or negative PD-Ll
mRNA
expression, including, but not limited to: pancreatic cancer (4% PD-L1+, Ayers
et al AACR
2015); prostate cancer (14% PD-L1+, Id.); triple negative breast cancer (29%
PD-Ll+, Id.);
melanoma (41% PD-Ll+, Id.); non-small cell lung cancer (42% PD-L1+, Id.);
urothelial
cancer (42% PD-Ll+, Id.); and head and neck cancer (59% PD-L1+, Id) (e.g.,
recurrent or
metastatic squamous cell carcinoma of the head and neck).
Methods. Uses and Medicaments
[0155] In one aspect, the invention relates to a method for treating cancer in
an individual
comprising administering to the individual a combination therapy which
comprises
pembrolizumab, a pembrolizumab variant or antigen-binding fragments thereof,
and
talimogene laherparepvec.
[0156] The combination therapy may also comprise one or more additional
therapeutic
agents. The additional therapeutic agent may be, e.g., a chemotherapeutic
agent, a
biotherapeu tic agent, an immunogenic agent (for example, attenuated cancerous
cells, tumor
antigens, antigen presenting cells such as dendritic cells pulsed with tumor
derived antigen or
nucleic acids, immune stimulating cytokines (for example, IL-2, IFNcx2, GM-
CSF), and cells
transfectecl with genes encoding immune stimulating cytokines such as but not
limited to
(3M-CSF). The specific dosage and dosage schedule of the additional
therapeutic agent can
further vary, and the optimal dose, dosing schedule and route of
administration will be
determined based upon the specific therapeutic agent that is being used.
38

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0157] Examples of chemotherapeutic agents include alkylating agents such as
thiotepa and
cyclosphosphamide; alkyl sulfonates such as busulfan, improsulfan and
piposulfan; aziri
dines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines
and
methylamelamines including altretamine, triethylenemelamine,
trietylenephosphoramide,
triethylenethiophosphoramide and trimethylolomelamine; acetogenins (especially
buliatacin
and bullatacinone); a camptothecin (including the synthetic analogue
topotecan); bryostatin;
cally statin; CC-1065 (including its adozelesin, carzelesin and bizelesin
synthetic analogues);
cryptophycins (particularly cryptophycin 1 and cryptophycin 8); dolastatin;
duocarmycin
(including the synthetic analogues, KW-2189 and CBI-TMI); eleutherobin;
pancrati statin; a
sarcodictyin; spongistatin; nitrogen mustards such as chlorambucil,
chlornaphazine,
cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine
oxide
hydrochloride, melphalan, novembichin, phenesterine, prednimustine,
trofosfamide, uracil
mustard; nitrosureas such as carmustine, chlorozotocin, fotemustine,
lomustine, nimustine,
ranimustine; antibiotics such as the enediyne antibiotics (e.g. calicheamicin,
especially
calicheamicin gammall and calicheamicin phill, see, e.g., Agnew, Chem. Intl.
Ed. Engl., 33 :
183-186 (1994); dplemicin, including dynemicin A; bisphosphonates, such as
clodronate; an
esperamicin; as well as neocaminostatin chromophore and related chromoprotein
enediyne
antibiotic chromomophores), aclacinomysins, actinomycin, authramycin,
azaserine,
bleomycins, cactinomycin, carabicin, caminomycin, carzinophilin, chromomycins,
dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine,
doxorubicin
(including morpholino- doxorubicin, cyanomorpholino-doxorubicin, 2-pyrrolino-
doxorubicin
and deoxydoxorubicin), epirubicin, esorubicin, idarubicin, marcellomycin,
mitomycins such
as mitomycin C, mycophenolic acid, nogalamycin, olivomycins, peplomycin,
potfiromycin,
puromycin, quelamycin, rodorubicin, streptonigrin, strepwwcin, tubercidin,
ubenimex,
zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-
fluorouracil (5-FU); folic
acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate;
purine analogs
such as fludarabine, 6- mercaptopurine, thiamiprine, thioguanine; pyrimidine
analogs such as
ancitabine, azacitidine, 6- azauridine, carmofur, cytarabine, dideoxyuridine,
doxifluridine,
enocitabine, floxuridine; androgens such as calusterone, dromostanolone
propionate,
epitiostanol, mepitiostane, testolactone; anti-adrenals such as
aminoglutethimide, mitotane,
trilostane; folic acid replenisher such as frolinic acid; aceglatone;
aldophosphamide
glycoside; aminolevulinic acid; eniluracil; amsacrine; bestrabucil;
bisantrene; edatraxate;
defofamine; demecolcine; diaziquone; elformithine; elliptinium acetate; an
epothilone;
39

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; maytansinoids
such as
maytansine and ansamitocins; mitoguazone; mitoxantrone; mopidamol; nitracrine;
pentostatin; phenamet; pirarubicin; losoxantrone; podophyllinic acid; 2-
ethylhydrazide;
procarbazine; razoxane; rhizoxin; sizofuran; spirogermanium; tenuazonic acid;
triaziquone; 2,
2',2"-trichlorotriethylamine; trichothecenes
(especially T-2 toxin, verracurin A. roridin A and anguidine); urethan;
vindesine;
dacarbazine; mannomu stifle; mitobronitol; mitolactol; pipobroman; gacytosine;
arabinoside
("Ara-C"); cyclophosphamide; thiotepa; taxoids, e.g. paclitaxel and doxetaxel;
chlorambucil;
gemcitabine;
6-thioguanine; mercaptopurine; methotrexate; platinum analogs such as
cisplatin and
carboplatin; vinblastine; platinum; etoposide (VP- 16); ifosfamide;
mitoxantrone; vincristine;
vinorelbine; novantrone; teniposide; edatrexate; daunomycin; aminopterin;
xeloda;
ibandronate; CPT-11; topoisomerase inhibitor RFS 2000; difluoromethylormthine
(DMF0);
retinoids such as retinoic acid; capecitabine; and pharmaceutically acceptable
salts, acids or
derivatives of any of the above. Also included are anti-hormonal agents that
act to regulate or
inhibit hormone action on tumors such as anti-estrogens and selective estrogen
receptor
modulators (SERMs), including, for example, tamoxifen. raloxifene,
droloxifene, 4-
hydroxytamoxifen, trioxifene, keoxifene,
LYI 17018, onapristone, and toremifene (Fareston); aromatase inhibitors that
inhibit the
enzyme aromatase, which regulates estrogen production in the adrenal glands,
such as, for
example, 4(5)- imidazoles, aminoglutethimide, megestrol acetate, exemestane,
formestane,
fadrozole, vorozole, letrozole, and anastrozole; and anti-androgens such as
flutamide,
nilutamide, bicalutamide, leuprolide, and goserelin; and pharmaceutically
acceptable salts,
acids or derivatives of any of the above.
[0158] Each therapeutic agent in a combination therapy of the invention may be
administered
either alone or in a medicament (also referred to herein as a pharmaceutical
composition)
which comprises the therapeutic agent and one or more pharmaceutically
acceptable carriers,
excipients and diluents, according to standard pharmaceutical practice.
[0159] Each therapeutic agent in a combination therapy of the invention may be
administered
simultaneously (i.e., in the same medicament), concurrently (i.e., in separate
medicaments
administered one right after the other in any order) or sequentially in any
order. Sequential
administration is particularly useful when the therapeutic agents in the
combination therapy
are in different dosage forms (one agent is a tablet or capsule and another
agent is a sterile

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
liquid) and/or are administered on different dosing schedules, e.g., a
chemotherapeutic that is
administered at least daily and a biotherapeutic that is administered less
frequently, such as
once weekly, once every two weeks, or once every three weeks and/or are
administered to
different parts of the body, e.g., one therapeutic agent is administered
intratumorally and one
therapeutic agent is administered systemically.
[0160] In particularly preferred embodiments, talimogene laherparepvec is
administered
before administration of pembrolizumab, a pembrolizumab variant or an antigen-
binding
fragment thereof. In other embodiments, talimogene laherparepvec is
administered after
administration of pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment
thereof. In another embodiment, talimogene laherparepvec is administered
concurrently with
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment thereof.
[0161] In some embodiments, at least one of the therapeutic agents in the
combination
therapy is administered using the same dosage regimen (dose, frequency and
duration of
treatment) that is typically employed when the agent is used as monotherapy
for treating the
same cancer. In other embodiments, the patient receives a lower total amount
of at least one
of the therapeutic agents in the combination therapy than when the agent is
used as
monotherapy, e.g., smaller doses, less frequent doses, and/or shorter
treatment duration.
[0162] In certain embodiments, talimogene laherparepvec is administered
intratumorally. In
certain embodiments, pembrolizumab, a pembrolizumab variant or an antigen-
binding
fragment thereof is administered parenterally.
[0163] A combination therapy of the invention may be used prior to or
following surgery to
remove a tumor and may be used prior to, during or after radiation therapy.
[0164] in some embodiments, a combination therapy of the invention is
administered to a
patient who has not been previously treated with a biotherapeu tic or
chemotherapeutic agent,
i.e., is cancer treatment-naive. In other embodiments, the combination therapy
is
administered to a patient who failed to achieve a sustained response after
prior therapy (e.g.,
after failed or ineffective therapy with a checkpoint inhibitor (e.g., with
anti-PD-Li therapy
or anti-PD-1 therapy (e.g., pembrolizumab, a pembrolizumab variant or an
antigen-binding
fragment thereof)), or a chemotherapeutic agent, i.e., is cancer treatment-
experienced.
[0165] A combination therapy of the invention is typically used to treat a
tumor that is large
enough to be found by palpation or by imaging techniques well known in the
art, such as
MRI, ultrasound, or CAT scan.
41

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0166] A combination therapy of the invention is preferably administered to a
human patient
who has a cancer that has a low CD8+ T-cell density, a negative 1FNy gene
signature, and/or
has a low or negative PD-L1 status.
[0167] Selecting a dosage regimen (also referred to herein as an
administration regimen) for
a combination therapy of the invention depends on several factors, including
the serum or
tissue turnover rate of the entity, the level of symptoms, the immunogenicity
of the entity,
and the accessibility of the target cells, tissue or organ in the individual
being treated.
Preferably, a dosage regimen maximizes the amount of each therapeutic agent
delivered to
the patient consistent with an acceptable level of side effects. Accordingly,
the dose amount
and dosing frequency of each biotherapeutic and chemotherapeutic agent in the
combination
depends in part on the particular therapeutic agent, the severity of the
cancer being treated,
and patient characteristics. Guidance in selecting appropriate doses of
antibodies, cytokines,
and small molecules are available. See, e.g., Wawrzynczak (1996) Antibody
Therapy, Bios
Scientific Pub. Ltd, Oxfordshire, UK; Kresina (ed.) (1991) Monoclonal
Antibodies,
Cytokines and Arthritis, Marcel Dekker, New York, NY; Bach (ed.) (1993)
Monoclonal
Antibodies and Peptide Therapy in Autoimmune Diseases, Marcel Dekker, New
York, NY;
Baert et al. (2003) New Engl. J. Med. 348:601-608; Milgrom et al (1999) New
Engl. J. Med.
341 : 1966-1973; Slamon et al. (2001) New Engl. J. Med. 344:783-792;
Beniaminovitz et al.
(2000) New Engl. J. Med. 342:613-619; Ghosh et al. (2003) New Engl. J. Med.
348:24-32;
Lipsky et al. (2000) New Engl. J. Med. 343 : 1594-1602; Physicians' Desk
Reference 2003
(Physicians' Desk Reference, 57th Ed); Medical Economics Company; ISBN:
1563634457;
57th edition (November 2002). Determination of the appropriate dosage regimen
may be
made by the clinician, e.g., using parameters or factors known or suspected in
the art to affect
treatment or predicted to affect treatment, and will depend, for example, the
patient's clinical
history (e.g., previous therapy), the type and stage of the cancer to be
treated and biomarkers
of response to one or more of the therapeutic agents in the combination
therapy. The optimal
dose for pembrolizumab in combination with talimogene laherparepvec may be
identified by
dose escalation or dose de-escalation of one or both of these agents.
[0168] The present invention also provides a medicament which comprises
pembrolizumab, a
pembrolizumab variant and/or an antigen-binding fragment thereof as described
above and a
pharmaceutically acceptable excipient. Pembrolizumab, a pembrolizumab variant
and/or an
antigen-binding fragment thereof may be produced in CHO cells using
conventional cell
culture and recovery/purification technologies.
42

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0169] In some embodiments, a medicament comprising pembrolizumab, a
pembrolizumab
variant and/or an antigen-binding fragment thereof may be provided as a liquid
formulation
or prepared by reconstituting a lyophilized powder with sterile water for
injection prior to
use. WO 2012/135408 describes the preparation of liquid and lyophilized
medicaments
comprising pembrolizumab that are suitable for use in the present invention.
In some
embodiments, a medicament comprising pembrolizumab is provided in a glass vial
which
contains about 100 mg of pembrolizumab in 4 ml of solution. Each 1 mL of
solution contains
25 mg of pembrolizumab and is formulated in: L-histidine (1.55 mg),
polysorbate 80 (0.2
mg), sucrose (70 mg), and water for injection, USP. The solution requires
dilution for IV
infusion.
[0170] Biotherapeutic agents in a combination therapy of the invention may be
administered
by continuous infusion, or by doses at intervals of, e.g., daily, every other
day, three times per
week, or one time each week, two weeks, three weeks, monthly, bimonthly, etc.
A total
weekly dose is generally at least 0.05 pg/kg, 0.2 g/kg, 0.5 pg/kg, 1 jig/kg,
10 jig/kg, 100
g/kg, 0.2 mg/kg, 1.0 mg/kg, 2.0 mg/kg, 10 mg/kg, 25 mg/kg, 50 mg/kg body
weight or
more. See, e.g., Yang et al. (2003) New Engl. J. Med. 349:427-434; Herold et
al. (2002)
New Engl. J. Med. 346: 1692-1698; Liu et al. (1999) J. Neurol. Neurosurg.
Psych. 67:451-
456; Portielji et al. (20003) Cancer Immunol. Immunother. 52: 133-144.
[0171] In certain embodiments that employ pembrolizumab, a pembrolizumab
variant and/or
an antigen-binding fragment thereof, the dosing regimen will comprise
administering
pembrolizumab, a pembrolizumab variant and/or an antigen-binding fragment
thereof at a
dose of 1, 2, 3, 5 or 10 mg/kg at intervals of about 14 days ( 2 days) or
about 21 days ( 2
days) or about 30 days ( 2 days) throughout the course of treatment. In a
preferred
embodiment, pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment
thereof is used at a dose of 2 mg/kg every 3 weeks (e.g., for melanoma). In
another preferred
embodiment, pembrolizumab, a pembrolizumab variant or an antigen-binding
fragment
thereof is used at a dose of 200 mg (fixed) every 3 weeks (e.g., for non-small
cell lung
cancer, head and neck squamous cell carcinoma, and/or Hodgkin's lymphoma).
[0172] In other embodiments that employ pembrolizumab, a pembrolizumab variant
and/or
an antigen-binding fragment thereof in the combination therapy, the dosing
regimen will
comprise administaing pembrolizumab, a pembrolizumab variant and/or an antigen-
binding
fragment thereof at a dose of from about 0.005 mg/kg to about 10 mg/kg, with
intra-patient
dose escalation. In other escalating dose embodiments, the interval between
doses will be
43

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
progressively shortened, e.g., about 30 days ( 2 days) between the first and
second dose,
about 14 days ( 2 days) between the second and third doses. In certain
embodiments, the
dosing interval will be about 14 days ( 2 days), for doses subsequent to the
second dose.
[0173] In certain embodiments, a subject will be administered a parenteral
dosing, e.g., an
intravenous (IV) infusion, of a medicament comprising any of pembrolizumab, a
pembrolizumab variant and/or an antigen-binding fragment thereof.
[0174] In a preferred embodiment of the invention, pembrolizumab, a
pembrolizumab variant
and/or an antigen-binding fragment thereof is administered in a liquid
medicament at a dose
selected from the group consisting of 1 mg/kg every two weeks (Q2W), 2 mg/kg
Q2W, 3
mg/kg Q2W, 5 mg/kg Q2W, 10 mg Q2W, 1 mg/kg every three weeks (Q3W), 2 mg/kg
Q3W,
3 mg/kg Q3W, 5 mg/kg Q3W, 10 mg Q3W and flat-dose equivalents of any of these
doses,
i.e., such as 200 mg Q3W. In some embodiments, pembrolizumab, a pembrolizumab
variant
and/or an antigen-binding fragment thereof is provided as a liquid medicament
which
comprises 25 mg/m1 pembrolizumab, 7% (w/v) sucrose, 0.02% (w/v) polysorbate 80
in 10
mM histidine buffer pH 5.5.
[0175] in some embodiments, the selected dose of pembrolizumab, a
pembrolizumab variant
and/or an antigen-binding fragment thereof is administered by IV infusion. In
one
embodiment, the selected dose of pembrolizumab, a pembrolizumab variant and/or
an
antigen-binding fragment thereof is administered by IV infusion over a time
period of
between 25 and 40 minutes, or about 30 minutes.
[0176] The present invention also provides a medicament which comprises
talimogene
laherparepvec and a pharmaceutically acceptable excipient. Talimogene
laherparepvec may
be suspended in a physiological buffer for intratumoral injection.
[0177] In certain embodiments, talimogene laherparepvec is administered by
intratumoral
injection into injectable tumors at a dose of up to 4.0 ml of 106 plaque
forming unithnL
(PFU/mL) at day 1 of week 1, followed by a dose of up to 4.0 ml of 108 PFU/mL
at day 1 of
weeks 4 and 6, and every 2 weeks ( 3 days) thereafter. The recommended volume
of
talimogene laherparepvec to be injected into the tumor(s) is dependent on the
size of the
tumor(s) and should be determined according to the injection volume guideline
in Table 1.
[0178] Table 1. Talimogene Laherparepvec Injection Volume Guidelines Based on
Tumor
Size
44

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Tumor Size (longest dimension) Maximum Injection Volume
> 5.0 cm 4.0 ml
> 2.5 cm to 5.0 cm 2.0m1
> 1.5 cm to 2.5 cm 1.0 ml
> 0.5 cm to 1.5 cm 0.5m1
< 0.5 cm 0.1 ml
[0179] All reasonably injectable lesions (cutaneous, subcutaneous and nodal
disease that can
be injected with or without ultrasound guidance) should be injected with the
maximum
dosing volume available on an individual dosing occasion. On each treatment
day,
prioritization of injections is recommended as follows: any new injectable
tumor that has
appeared since the last injection; by tumor size, beginning with the largest
tumor; any
previously uninjectable tumor(s) that is now injectable. The compositions may
comprise one
or more substances selected from the group consisting of a buffer, an
antioxidant such as
ascorbic acid, a low molecular weight polypeptide (such as those having fewer
than 10 amino
acids), a protein, an amino acid, a carbohydrate such as glucose, sucrose or
dextrins, a
chelating agent such as EDTA, glutathione, a stabilizer, and an excipient.
Neutral buffered
saline or saline mixed with specific serum albumin are examples of appropriate
diluents. In
accordance with appropriate industry standards, preservatives such as benzyl
alcohol may
also be added. The composition may be formulated as a lyophilizate using
appropriate
excipient solutions (e.g., sucrose) as diluents. Suitable components are
nontoxic to recipients
at the dosages and concentrations employed.
[0180] In some embodiments, the patient is selected for treatment with the
combination
therapy of the invention if the patient has: (1) a CD8+ T-celi infiltration
density of fewer than
about 1500, about 1400, about 1300, about 1200, about 1100, about 1000, about
900, about
800, about 700, about 600, or about 500 cells per 1 mm2 sample (or per 1 mL
sample), a low
or negative PD-L1 status, and/or a negative IFNy gene signature; and (2) has
been diagnosed
with melanoma (e.g., cutaneous, metastatic, uveal), non-small cell lung
cancer, head and
neck cancer (e.g., recurrent or metastatic squamous cell carcinoma of the head
and neck),
colorectal cancer, breast cancer (e.g., HER2+, HER2- (HR+), triple-negative),
ovarian cancer,
bladder cancer (e.g., transitional cell cancer, urothelial cancer), prostate
cancer (e.g.,
castration-resistant), sarcoma (e.g., soft tissue, bone), renal. cell cancer,
gastric cancer,
esophageal cancer, anal canal cancer, biliary tract cancer or pancreatic
cancer.

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0181] In other embodiments, the patient is selected for treatment with the
combination
therapy of the invention if the patient has: (1) a cancer that is responsive
to monotherapy with
a checkpoint inbibitor (e.g., with anti-PD-Li therapy or anti-PD-1 therapy
(e.g.,
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)); (2)
which may optionally be associated with one or more of a CD8+ T-cell
infiltration density of
fewer than about 1500, about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells per 1 mm2 sample (or
per 1 mL
sample), a low or negative PD-L1 status and a negative IFNy gene signature;
and (3) has been
diagnosed with melanoma (e.g., cutaneous, metastatic, uveal), lung cancer
(e.g., non-small
cell lung cancer, small cell lung cancer), head and neck cancer (e.g.,
recurrent or metastatic
squamous cell carcinoma of the head and neck, nasopharyngeal cancer, thyroid
cancer,
salivary cancer, esophageal cancer), breast cancer (e.g., ER+/FIER2- breast
cancer,
inflammatory, triple-negative breast cancer), ovarian cancer, cervical cancer,
bladder cancer
(e.g., urothelial cancer), renal cell cancer, gastrointestinal cancer (e.g.,
hepatocellular cancer,
colorectal cancer, anal cancer), biliary tract cancer, multiple myeloma,
lymphoma (e.g.,
mediastinal large B-cell lymphoma, Hodgkin's lymphoma) or mesothelioma.
[0182] The medicaments described herein may be provided as a kit which
comprises a first
container and a second container and a package insert. The first container
contains at least
one dose of a medicament comprising a pembrolizumab, a pembrolizumab variant
and/or an
antigen-binding fragment thereof, and the second container contains at least
one dose of
talimogene laherparepvec. The kit can optionally comprise a package insert, or
label, which
includes instructions for treating a patient for cancer using the medicaments.
The first and
second containers may be comprised of the same or different shapes (e.g.,
vials, syringes and
bottles) and/or materials (e.g., plastic or glass). The kit may further
comprise other materials
that may be useful in administering the medicaments, such as diluents,
filters, IV bags and
lines, needles and syringes. In some preferred embodiments of the kit, the
instructions state
that the medicaments are intended for use in treating a patient having a
cancer that has a
CD8+ T-cell density of fewer than 1,000 cells/mm2, a negative IFNy gene
signature and/or a
low or negative PD-Li status.
46

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Pharmaceutical Compositions
[0183] The invention pertains to uses of the above-described agents for
prophylactic and/or
therapeutic treatments as described Infra. Accordingly, pembrolizumab, the
pembrolizumab
variant and/or the antigen-binding fragment thereof and/or talimogene
laherparepvec of the
present invention can be incorporated into pharmaceutical compositions
suitable for
administration. Such compositions typically comprise pembrolizumab, the
pembrolizumab
variant and/or the antigen-binding fragment thereof or talimogene
laherparepvec and a
pharmaceutically acceptable carrier. As used herein the language
"pharmaceutically
acceptable carrier" is intended to include any and all solvents, dispersion
media, coatings,
antibacterial and antifungal agents, isotonic and absorption delaying agents,
and the like,
compatible with pharmaceutical administration. The use of such media and
agents for
pharmaceutically active substances is well known in the art. Except insofar as
any
conventional media or agent is incompatible with the active compound, use
thereof in the
compositions is contemplated. Supplementary active compounds can also be
incorporated
into the compositions.
[0184] A pharmaceutical composition of the invention is formulated to be
compatible with its
intended route of administration. Examples of routes of administration include
parenteral,
e.g., intravenous, intradermal, subcutaneous, intraperitoneal, intramuscular,
transdermal
(topical), and transmucosal administration. Solutions or suspensions used for
parenteral,
intradermal, or subcutaneous application can include the following components:
a sterile
diluent such as water for injection, saline solution, fixed oils, polyethylene
glycols, glycerine,
propylene glycol or other synthetic solvents; antibacterial agents such as
benzyl alcohol or
methyl paraberis; antioxidants such as ascorbic acid or sodium bisulfite;
chelating agents such
as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or
phosphates and agents
for the adjustment of tonicity such as sodium chloride or dextrose. pH can be
adjusted with
acids or bases, such as hydrochloric acid or sodium hydroxide. The parenteral
preparation
can be enclosed in ampoules, disposable syringes or multiple dose vials made
of glass or
plastic.
[0185] Pharmaceutical compositions suitable for injectable use include sterile
aqueous
solutions (where water soluble) or dispersions and sterile powders for the
extemporaneous
preparation of sterile injectable solutions or dispersion. For intravenous,
IS, ICV and/or IT
administration, suitable carriers include physiological saline, bacteriostatic
water, Cremophor
ELTM (BASF, Parsippany, N.J.) or phosphate buffered saline (PBS). In all
cases, the
47

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
composition must be sterile and should be fluid to the extent that easy
syringability exists. It
must be stable under the conditions of manufacture and storage and must be
preserved against
the contaminating action of microorganisms such as bacteria and fungi. The
carrier can be a
solvent or dispersion medium containing, for example, water, ethanol, polyol
(for example,
glycerol, propylene glycol, and liquid polyethylene glycol, and the like), and
suitable
mixtures thereof. The proper fluidity can be maintained, for example, by the
use of a coating
such as lecithin, by the maintenance of the required particle size in the case
of dispersion and
by the use of surfactants. Prevention of the action of microorganisms can be
achieved by
various antibacterial and antifungal agents, for example, parabens,
chlorobutanol, phenol,
ascorbic acid, thimerosal, and the like. In many cases, it will be preferable
to include isotonic
agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium
chloride in the
composition. Prolonged absorption of the injectable compositions can be
brought about by
including in the composition an agent which delays absorption, for example,
aluminum
monostearate and gelatin.
[0186] Sterile injectable solutions can be prepared by incorporating the
active compound in
the required amount in an appropriate solvent with one or a combination of
ingredients
enumerated above, as required, followed by filtered sterilization. Generally,
dispersions are
prepared by incorporating the active compound into a sterile vehicle which
contains a basic
dispersion medium and the required other ingredients from those enumerated
above. In the
case of sterile powders for the preparation of sterile injectable solutions,
the preferred
methods of preparation are vacuum drying and freeze-drying which yields a
powder of the
active ingredient plus any additional desired ingredient from a previously
sterile-filtered
solution thereof.
[0187] Systemic administration can be by transmucosal or transdermal means.
For
transmucosal or transdermal administration, penetrants appropriate to the
barrier to be
permeated are used in the formulation. Such penetrants are generally known in
the art, and
include, for example, for transmucosal administration, detergents, bile salts,
and fusidic acid
derivatives. Transmucosal administration can be accomplished through the use
of nasal
sprays or suppositories. For transdermal administration, the active compounds
are
formulated into ointments, salves, gels, or creams as generally known in the
art.
[0188] In one embodiment, pembrolizumab, a pembrolizumab variant and/or an
antigen-
binding fragment thereof or talimogene laherparepvec are prepared with
carriers that will
protect the compound against rapid elimination from the body, such as a
controlled release
48

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
formulation, including implants and microencapsulated delivery systems.
Biodegradable,
biocompatible polymers can be used, such as ethylene vinyl acetate,
polyanhydrides,
polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Methods for
preparation of
such formulations will be apparent to those skilled in the art. The materials
can also be
obtained commercially from Alza Corporation and Nova Pharmaceuticals, Inc.
Liposomal
suspensions (including liposomes targeted to infected cells with monoclonal
antibodies to
viral antigens) can also be used as pharmaceutically acceptable carriers.
These can be
prepared according to methods blown to those skilled in the art, for example,
as described in
U.S. Pat. No. 4522,811.
[0189] It is especially advantageous to formulate parenteral compositions in
dosage unit form
for ease of administration and uniformity of dosage. Dosage unit form as used
herein refers
to physically discrete units suited as unitary dosages for the subject to be
treated; each unit
containing a predetermined quantity of active compound calculated to produce
the desired
therapeutic effect in association with the required pharmaceutical carrier.
The specification
for the dosage unit forms of the invention are dictated by and directly
dependent on the
unique characteristics of the active compound and the particular therapeutic
effect to be
achieved, and the limitations inherent in the art of compounding such an
active compound for
the treatment of individuals.
[0190] The pharmaceutical compositions can be included in a container, pack or
dispenser
together with optional instructions for administration.
[0191] The pharmaceutical compositions of the present invention may be
administered in a
number of ways depending upon whether local or systemic treatment is desired
and upon the
area to be treated. Administration may be intratumoral or parenteral.
Parenteral
administration includes intravenous drip, subcutaneous, intraperitoneal or
intramuscular
injection, intrathecal, or intraventricular administration.
[0192] Formulations for parenteral administration may include sterile aqueous
solutions
which may also contain buffers, diluents and other suitable additives.
Intraventricular
injection may be facilitated by an intraventricular catheter, for example,
attached to a
reservoir. For intravenous use, the total concentration of solutes should be
controlled to
render the preparation isotonic.
[0193] The types of pharmaceutical excipients that are useful as carriers
include stabilizers
such as human serum albumin (HSA), bulking agents such as carbohydrates, amino
acids and
49

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
polypeptides; pH adjusters or buffers; salts such as sodium chloride; and the
like. These
carriers may be in a crystalline or amorphous form or may be a mixture of the
two.
[0194] Bulking agents that are particularly valuable include compatible
carbohydrates,
polypeptides, amino acids or combinations thereof. Suitable carbohydrates
include
monosaccharides such as galactose, D-mannose, sorbose, and the like;
disaccharides, such as
lactose, trehalose, and the like; cyclodextrins, such as 2-hydroxypropyl.beta.-
cyclodextrin;
and polysaccharides, such as raffinose, maltodextrins, dextrans, and the like;
alditols, such as
mannitol, xylitol, and the like. A preferred group of carbohydrates includes
lactose,
trehalose, raffinose maltodextrins, and mannitol. Suitable polypeptides
include aspartame.
Amino acids include alanine and glycine, with glycine being preferred.
[0195] Suitable pH adjusters or buffers include organic salts prepared from
organic acids and
bases, such as sodium citrate, sodium ascorbate, and the like; sodium citrate
is preferred.
[0196] In one embodiment, unit doses or measured doses of a composition that
include the
pembrolizumab, pembrolizumab variant and/or antigen-binding fragment thereof
or
talimogene laherparepvec are dispensed by an implanted device. The device can
include a
sensor that monitors a parameter within a subject. For example, the device can
include a
pump, such as an osmotic pump and, optionally, associated electronics.
Biomarker Testing
[0197] A biomarker expression score (e.g., an IFNI gene signature score and/or
PD-Li
status) can be determined in a sample of tumor tissue removed from a subject.
The tumor
may be primary or recurrent, and may be of any type (as described above), any
stage (e.g.,
Stage I, II, III, or IV or an equivalent of other staging system), and/or any
histology. The
subject may be of any age, gender, treatment history and/or extent and
duration of remission.
[0198] The tumor sample can be obtained by a variety of procedures including,
but not
limited to, surgical excision, aspiration or biopsy. The tissue sample may be
sectioned and
assayed as a fresh specimen. Alternatively, the tissue sample may be frozen
for further
sectioning. In some preferred embodiments, the tissue sample is preserved by
fixing and
embedding in paraffin or the like.
[0199] The tumor tissue sample may be fixed by conventional methodology, with
the length
of fixation depending on the size of the tissue sample and the fixative used.
Neutral buffered
formalin, glutaraldehyde, Bouin's and paraformaldehyde are non-limiting
examples of
fixatives. In preferred embodiments, the tissue sample is fixed with formalin.
In some

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
embodiments, the fixed tissue sample is also embedded in paraffin to prepare a
formalin-
fixed paraffin-embedded (FFPE) tissue sample.
[0200] Typically, the tissue sample is fixed and dehydrated through an
ascending series of
alcohols, infiltrated and embedded with paraffin or other sectioning media so
that the tissue
sample may be sectioned. Alternatively, the tumor tissue sample is first
sectioned and then
the individual sections are fixed.
[0201] In some preferred embodiments, a biomarker expression score for a tumor
is
determined using FFPE tissue sections of about 3-5 micrometers, and preferably
4 or 5
micrometers, which are mounted and dried on a microscope slide.
Diagnostic Testing for IFN-y Gene Signature Score and/or PD-Li Status
[0202] In one embodiment, the tested tumor sample is from a cancer associated
with a low or
negative PD-L1 status and/or a negative IFNy gene signature, and optionally a
CD8+ T-cell
infiltration density of fewer than about 1500, about 1400, about 1300, about
1200, about
1100, about 1000, about 900, about 800, about 700, about 600, or about 500
cells per 1 mm2
sample (or per I mL sample), including, but not limited to, melanoma (e.g.,
cutaneous,
metastatic, uveal), non-small cell lung cancer, head and neck cancer (e.g.,
recurrent or
metastatic squamous cell carcinoma of the head and neck), colorectal cancer,
breast cancer
(e.g., HER2+, HER2- (HR+), triple-negative), ovarian cancer, bladder cancer
(e.g.,
transitional cell cancer, urothelial cancer), prostate cancer (e.g.,
castration-resistant), sarcoma
(e.g., soft tissue, bone), renal cell cancer, gastric cancer, esophageal
cancer, anal canal cancer,
biliary tract cancer or pancreatic cancer.
[0203] In another embodiment, the tested tumor sample is from a cancer that is
responsive to
monotherapy with a checkpoint inbibitor (e.g., with anti-PD-Li therapy or anti-
PD-1 therapy
(e.g., pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)),
which may optionally be associated with one or more of a CD8+ T-cell
infiltration density of
fewer than about 1500. about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells per 1 mm2 sample (or
per 1 mL
sample), a low or negative PD-Ll status and a negative IFNy gene signature,
including, but
not limited to, melanoma (e.g., cutaneous, metastatic, uveal), lung cancer
(e.g., non-small cell
lung cancer, small cell lung cancer), head and neck cancer (e.g., recurrent or
metastatic
squamous cell carcinoma of the head and neck, nasopharyngeal cancer, thyroid
cancer,
salivary cancer, esophageal cancer), breast cancer (e.g., ER+/HER2- breast
cancer, triple-
negative breast cancer), ovarian cancer, cervical cancer, bladder cancer
(e.g., urothelial
51

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
cancer), renal cell cancer, gastrointestinal cancer (e.g., hepatocellular
cancer, colorectal
cancer, anal cancer), biliary tract cancer, multiple myeloma, lymphoma (e.g.,
mediastinal
large B-cell lymphoma, Hodgkin's lymphoma) or mesothelioma.
[0204] Once a suitable sample of tumor tissue has been obtained, it can be
analyzed to
quantitate the expression level of each of the genes that comprise the
particular IFNI, gene
signature to be scored, e.g., one or any combination of IFNy, STAT1, CCR5,
CXCL9, PRF1,
HLA-DRA, CXCL10, CXCL11, IDO1 and GZMA, or the mRNA levels of PD-Li. The
phrase "determine the expression level of a gene" as used herein refers to
detecting and
quantifying RNA transcribed from that gene or a protein translated from such
RNA. The
term "RNA transcript" includes mRNA transcribed from the gene, and/or specific
spliced
variants thereof and/or fragments of such mRNA and spliced variants. In
preferred
embodiments, the RNA transcripts whose expression are measured are the
transcripts in
Figure 11 and/or PD-Li transcripts.
[0205] A person skilled in the art will appreciate that a number of methods
can be used to
isolate RNA from the tissue sample for analysis. For example, RNA may be
isolated from
frozen tissue samples by homogenization in guanidinium isothiocyanate and acid
phenol-
chloroform extraction. Commercial kits are available for isolating RNA from
FFPE samples.
[0206] Persons skilled in the art are also aware of several methods useful for
detecting and
quantifying the level of RNA transcripts within the isolated RNA or whole cell
lysates.
Quantitative detection methods include, but are not limited to, arrays (i.e.,
microarrays),
quantitative real time PCR (RT-PCR), multiplex assays, nuclease protection
assays, and
Northern blot analyses. Generally, such methods employ labeled probes that are
complimentary to a portion of each transcript to be detected. Probes for use
in these methods
can be readily designed based on the known sequences of the genes and the
transcripts
expressed thereby. In some preferred embodiments, the probes are designed to
hybridize to
each of the gene signature transcripts identified in Figure 11. Suitable
labels for the probes
are well-known and include, e.g., fluorescent, chemilumineseent and
radioactive labels.
[0207] in some embodiments, assaying a tumor sample for a gene signature of
the invention
employs detection and quantification of RNA levels in real-time using nucleic
acid sequence
based amplification (NASBA) combined with molecular beacon detection
molecules.
NASBA is described, e.g., in Compton J., Nature 350 (6313)91-92 (1991). NASBA
is a
single-step isothermal RNA-specific amplification method. Generally, the
method involves
the following steps: RNA template is provided to a reaction mixture, where the
first primer
52

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
attaches to its complementary site at the 3' end of the template; reverse
transcriptase
synthesizes the opposite, complementary DNA strand; RNase H destroys the RNA
template
(RNase H only destroys RNA in RNA-DNA hybrids, but not single-stranded RNA);
the
second primer attaches to the 3' end of the DNA strand, and reverse
transcriptase synthesizes
the second strand of DNA; and 17 RNA polymerase binds double-stranded DNA and
produces a complementary RNA strand which can be used again in step 1, such
that the
reaction is cyclic.
[0208] In other embodiments, the assay format is a flap endonuclease-based
format, such as
the InvaderTM assay (Third Wave Technologies). In the case of using the
invader method, an
invader probe containing a sequence specific to the region 3' to a target
site, and a primary
probe containing a sequence specific to the region 5' to the target site of a
template and an
unrelated flap sequence, are prepared. Cleavase is then allowed to act in the
presence of
these probes, the target molecule, as well as a FRET probe containing a
sequence
complementary to the flap sequence and an auto-complementary sequence that is
labeled with
both a fluorescent dye and a quencher. When the primary probe hybridizes with
the template,
the 3' end of the invader probe penetrates the target site, and this structure
is cleaved by the
Cleavase resulting in dissociation of the flap. The flap binds to the FRET
probe and the
fluorescent dye portion is cleaved by the Cleavase resulting in emission of
fluorescence.
[0209] In yet other embodiments, the assay format employs direct mRNA capture
with
branched DNA (QuantiGenerm, Panomics) or Hybrid Capture' (Digene).
[0210] One example of an array technology suitable for use in measuring
expression of the
genes in an IFN-y gene signature or measuring expression of PD-Ll is the
ArrayPlateTM
assay technology sold by HTG Molecular, Tucson AZ, and described in Martel, R.
R., et al.,
Assay and Drug Development Technologies 1(1):61-71, 2002. In brief, this
technology
combines a nuclease protection assay with array detection. Cells in microplate
wells are
subjected to a nuclease protection assay. Cells are lysed in the presence of
probes that bind
targeted mRNA species. Upon addition of Si nuclease, excess probes and
unhybridized
mRNA are degraded, so that only mRNA : probe duplexes remain. Alkaline
hydrolysis
destroys the mRNA component of the duplexes, leaving probes intact. After the
addition of a
neutralization solution, the contents of the processed cell culture plate are
transferred to
another ArrayPlateTM called a programmed ArrayPlateTM. ArrayPlatesTM contain a
16-
element array at the bottom of each well. Each array element comprises a
position-specific
anchor oligonucleotide that remains the same from one assay to the next. The
binding
53

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
specificity of each of the 16 anchors is modified with an oligonucleotide,
called a
programming linker oligonucleotide, which is complementary at one end to an
anchor and at
the other end to a nuclease protection probe. During a hybridization reaction,
probes
transferred from the culture plate are captured by immobilized programming
linker.
Captured probes are labeled by hybridization with a detection linker
oligonucleotide, which is
in turn labeled with a detection conjugate that incorporates peroxidase. The
enzyme is
supplied with a chemiluminescent substrate, and the enzyme-produced light is
captured in a
digital image. Light intensity at an array element is a measure of the amount
of
corresponding target mRNA present in the original cells.
[0211] By way of further example, DNA microarrays can be used to measure gene
expression. In brief, a DNA microarray, also referred to as a DNA chip, is a
microscopic
array of DNA fragments, such as synthetic oligonucleotides, disposed in a
defined pattern on
a solid support, wherein they are amenable to analysis by standard
hybridization methods (see
Schena, BioEssays 18:427 (1996)). Exemplary microarrays and methods for their
manufacture and use are set forth in T. R. Hughes et al., Nature Biotechnology
9:342-347
(2001). A number of different microarray configurations and methods for their
production
are known to those of skill in the art and are disclosed in U.S. Pat. Nos.
5,242,974; 5,384,261;
5,405,783; 5,412,087; 5,424,186; 5,429,807; 5,436,327; 5,445,934; 5,556,752;
5,405,783;
5,412,087; 5,424,186; 5,429,807; 5,436,327; 5,472,672; 5,527,681; 5,529,756;
5,545,531;
5,554,501; 5,561,071; 5,571,639; 5,593,839; 5,624,711; 5,700,637; 5,744,305;
5,770,456;
5,770,722; 5,837,832; 5,856,101; 5,874,219; 5,885,837; 5,919,523; 6,022,963;
6,077,674;
and U.S. Pat. No. 6,156,501; Shena, et al., Tibtech 6:301-306, 1998; Duggan,
et al., Nat.
Genet. 2:10-14, 1999; Bowtell, et al., Nat. Genet. 21:25-32, 1999; Lipshutz,
et al., Nat.
Genet. 21:20-24, 1999; Blanchard, et al., Biosensors and Bioelecu-onics 77:687-
90, 1996;
Maskos, et al., Nucleic Acids Res. 2:4663-69, 1993; and Hughes, et al., Nat.
Biotechnol.
79:342-347, 2001. Patents describing methods of using arrays in various
applications include:
U.S. Pat. Nos. 5,143,854; 5,288,644; 5,324,633; 5,432,049; 5,470,710;
5,492,806; 5,503,980;
5,510,270; 5,525,464; 5,547,839; 5,580,732; 5,661,028; 5,848,659; and
5,874,219; the
disclosures of which are herein incorporated by reference in their entireties
for all purposes.
[0212] in one embodiment, an array of oligonucleotides may be synthesized on a
solid
support. Exemplary solid supports include glass, plastics, polymers, metals,
metalloids,
ceramics, organics, etc. Using chip masking technologies and photoprotective
chemistry, it is
possible to generate ordered arrays of nucleic acid probes. These arrays,
which are known,
54

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
for example, as "DNA chips" or very large scale immobilized polymer arrays
("VLSIPS@"
arrays), may include millions of defined probe regions on a substrate having
an area of about
1 cm2 to several cm2, thereby incorporating from a few to millions of probes
(see, e.g., U.S.
Pat. No. 5,631,734).
[0213] To compare expression levels, labeled nucleic acids may be contacted
with the array
under conditions sufficient for binding between the target nucleic acid and
the probe on the
array. In one embodiment, the hybridization conditions may be selected to
provide for the
desired level of hybridization specificity; that is, conditions sufficient for
hybridization to
occur between the labeled nucleic acids and probes on the microarray.
[0214] Hybridization may be carried out in conditions permitting essentially
specific
hybridization. The length and GC content of the nucleic acid will determine
the thermal
melting point and thus, the hybridization conditions necessary for obtaining
specific
hybridization of the probe to the target nucleic acid. These factors are well-
known to a
person of skill in the art, and may also be tested in assays. An extensive
guide to nucleic acid
hybridization may be found in Tijssen, et al. (Laboratory Techniques in
Biochemistry and
Molecular Biology, Vol. 24: Hybridization With Nucleic Acid Probes, P.
Tijssen, ed.;
Elsevier, N.Y. (1993)). The methods described above will result in the
production of
hybridization patterns of labeled target nucleic acids on the array surface.
The resultant
hybridization patterns of labeled nucleic acids may be visualized or detected
in a variety of
ways, with the particular manner of detection selected based on the particular
label of the
target nucleic acid. Representative detection means include scintillation
counting,
autoradiography, fluorescence measurement, calorimetric measurement, light
emission
measurement, light scattering, and the like.
[0215] One such method of detection utilizes an array scanner that is
commercially available
(Affymetrix, Santa Clara, CA), for example, the 417 Arrayer, the 418 Array
Scanner, or
the Agilent Gene Array Scanner. This scanner is controlled from a system
computer with
an interface and easy-to-use software tools. The output may be directly
imported into or
directly read by a variety of software applications. Exemplary scanning
devices are described
in, for example, U.S. Pat. Nos. 5,143,854 and 5,424,186.
[0216] A preferred assay method to measure biomarker transcript abundance
includes using
the nCounter Analysis System marketed by NanoString@ Technologies (Seattle,
Wash.
USA). This system, which is described by Geiss et al., Nature Biotechnol.
2(3):317-325
(2008), utilizes a pair of probes, namely, a capture probe and a reporter
probe, each

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
comprising a 35- to 50-base sequence complementary to the transcript to be
detected. The
capture probe additionally includes a short common sequence coupled to an
immobilization
tag, e.g. an affinity tag that allows the complex to be immobilized for data
collection. The
reporter probe additionally includes a detectable signal or label. e.g. is
coupled to a color-
coded tag. Following hybridization, excess probes are removed from the sample,
and
hybridized probe/target complexes are aligned and immobilized via the affinity
or other tag in
a cartridge. The samples are then analyzed, for example using a digital
analyzer or other
processor adapted for this purpose. Generally, the color-coded tag on each
transcript is
counted and tabulated for each target transcript to yield the expression level
of each transcript
in the sample. This system allows measuring the expression of hundreds of
unique gene
transcripts in a single multiplex assay using capture and reporter probes
designed by Nano-
String.
[0217] In measuring expression of the genes in an IFN-y gene signature or to
determine Pll-
Ll status described herein, the absolute expression of each of the genes in a
tumor sample is
compared to a control. For example, the control can be the average level of
expression of
each of the genes, respectively, in a pool of subjects. To increase the
sensitivity of the
comparison, however, the expression level values are preferably transformed in
a number of
ways.
[0218] For example, the expression level of each gene in the gene signature
can be
normalized by the average expression level of all of the genes, or the total
expression level of
all genes. the expression level of which is determined, or by the average
expression level of a
set of control genes. Thus, in one embodiment, the genes are represented by a
set of probes,
and the expression level of each of the genes is normalized by the mean or
median expression
level across all of the genes represented, including any genes that are not
part of the gene
signature of interest. In a specific embodiment, the normalization is carried
out by dividing
the median or mean level of expression of all of the genes on the microarray.
In another
embodiment, the expression levels of the signature genes are normalized by the
mean or
median level of expression of a set of control genes. In a specific
embodiment, the control
genes comprise housekeeping genes. In another specific embodiment, the
normalization is
accomplished by dividing by the median or mean expression level of the control
genes.
[0219] The sensitivity of a gene signature score will also be increased if the
expression levels
of individual genes in the gene signature are compared to the expression of
the same genes in
a pool of tumor samples. Preferably, the comparison is to the mean or median
expression
56

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
level of each signature gene in the pool of samples. Such a comparison may be
accomplished, for example, by dividing by the mean or median expression level
of the pool
for each of the genes from the expression level each of the genes in the
subject sample of
interest. This has the effect of accentuating the relative differences in
expression between
genes in the sample and genes in the pool as a whole, making comparisons more
sensitive
and more likely to produce meaningful results than the use of absolute
expression levels
alone. The expression level data may be transformed in any convenient way.
Preferably, the
expression level data for all is log transformed before means or medians are
taken.
[0220] In performing comparisons to a pool, two approaches may be used. First,
the
expression levels of the signature genes in the sample may be compared to the
expression
level of those genes in the pool, where nucleic acid derived from the sample
and nucleic acid
derived from the pool are hybridized during the course of a single experiment.
Such an
approach requires that a new pool of nucleic acid be generated for each
comparison or limited
numbers of comparisons, and is therefore limited by the amount of nucleic acid
available.
Alternatively, and preferably, the expression levels in a pool, whether
normalized and/or
transformed or not, are stored on a computer, or on computer-readable media,
to be used in
comparisons to the individual expression level data from the sample (i.e.,
single-channel
data).
f02211 When comparing a subject's tumor sample with a standard or control, the
expression
value of a particular gene in the sample is compared to the expression value
of that gene in
the standard or control. For each gene in a gene signature of the invention,
the log to ratio is
created for the expression value in the individual sample relative to the
standard or control.
A score for an IFN-y gene signature or PD-Li expression is calculated by
determining the
mean log(10) ratio of the genes in the signature. If the gene signature score
for the test
sample is above a pre-determined threshold for that gene signature, then the
sample is
considered to be positive for an IFN-y gene signature biomarker. In one
embodiment of the
invention, the pre-determined threshold is set at any number between 2.17 and
2.69 (i.e.,
2.18, 2.19, 2.20. . . 2.66, 2.67, 2.68). The pre-determined threshold may also
be the mean,
median, or a percentile of scores for that gene signature in a collection of
samples or a pooled
sample used as a standard or control.
[0222] It will be recognized by those skilled in the art that other
differential expression
values, besides log(10) ratio, may be used for calculating a signature score,
as long as the
value represents an objective measurement of transcript abundance of the
genes. Examples
57

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
include, but are not limited to: xdev, error-weighted log (ratio), and mean
subtracted
log(intensity).
[0223] in one preferred embodiment, raw expression values are normalized by
performing
quantile normalization relative to the reference distribution and subsequent
log 10-
transformation. When the gene expression is detected using the nCounter
Analysis System
marketed by NanoString , NanoString Technologies, the reference distribution
is generated
by pooling reported (i.e., raw) counts for the test sample and one or more
control samples
(preferably at least 2 samples, more preferably at least any of 4, 8 or 16
samples) after
excluding values for technical (both positive and negative control) probes and
without
performing intermediate normalization relying on negative (background-
adjusted) or positive
(synthetic sequences spiked with known titrations). The IFNI' signature score
is then
calculated as the arithmetic mean of normalized values for each of the genes
in the gene
signature, e.g., one or any combination of STAT1, CCR5, CXCL9, PRF1, and HLA-
DRA or
each of 1FNy, STAT1, CCR5, CXCL9, PRF1, HLA-DRA, CXCL10, CXCL11, IDO1 and
GZMA, or of PD-Li.
[0224] In some preferred embodiments, the reference distribution is generated
from raw
expression counts for a normalization set of genes, which consists essentially
of each of the
genes in the set of 400 genes listed in Figure 11, or a subset thereof. The
subset may consist
of at least any of 25, 50, 75, 100, 125, 150, 175, 200, 225, 250, 275, 300,
325, 350, 375 or
any whole number in between 25 and 400.
[0225] Each of the steps of obtaining a tissue sample, preparing one or more
tissue sections
therefrom for a gene signature biomarker assay, performing the assay, and
scoring the results
may be performed by separate individuals/entities at separate locations. For
example, a
surgeon may obtain by biopsy a tissue sample from a cancer patient's tumor and
then send the
tissue sample to a pathology lab, which may fix the tissue sample and then
prepare one or
more slides, each with a single tissue section, for the assay. The slide(s)
may be assayed soon
after preparation, or stored for future assay. The lab that prepared a tissue
section may
conduct the assay or send the slide(s) to a different lab to conduct the
assay. A pathologist or
trained professional who scores the slide(s) for an IFNI, gene signature may
work for the
diagnostic lab, or may be an independent contractor. Alternatively, a single
diagnostic lab
obtains the tissue sample from the subject's physician or surgeon and then
performing all of
the steps involved in preparing tissue sections, assaying the slide(s) and
calculating the gene
signature score for the tissue section(s).
58

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0226] In some embodiments, the individuals involved with preparing and
assaying the tissue
section for a gene signature biomarker do not know the identity of the subject
whose sample
is being tested; i.e., the sample received by the laboratory is made anonymous
in some
manner before being sent to the laboratory. For example, the sample may be
merely
identified by a number or some other code (a "sample ID") and the results of
the assay are
reported to the party ordering the test using the sample ID. In preferred
embodiments, the
link between the identity of a subject and the subject's tissue sample is
known only to the
individual or to the individual's physician.
[0227] In some embodiments, after the test results have been obtained, the
diagnostic
laboratory generates a test report, which may comprise any one or both of the
following
results: the tissue sample was biomarker positive or negative, the gene
signature score for the
tumor sample and the reference score for that gene signature. The test report
may also
include a list of genes whose expression was analyzed in the assay.
[0228] In other embodiments, the test report may also include guidance on how
to interpret
the results for predicting if a subject is likely to respond to a
pembrolizumab, pembrolizumab
variant and/or antigen-binding fragment thereof / talimogene laherparepvec
combination
therapy. For example, in one embodiment, the tested tumor sample is from a
cancer
including, but not limited to, melanoma (e.g., cutaneous, metastatic, uveal),
non-small cell
lung cancer, head and neck cancer (e.g., recurrent or metastatic squamous cell
carcinoma of
the head and neck,), colorectal cancer, breast cancer (e.g., HER2+, HER2-
(HR+), triple-
negative), ovarian cancer, bladder cancer (e.g., transitional cell cancer,
urothelial cancer),
prostate cancer (e.g., castration-resistant), sarcoma (e.g., soft tissue,
bone), renal cell cancer,
gastric cancer, esophageal cancer, anal canal cancer, biliary tract cancer or
pancreatic cancer
and has a negative IFN-y gene signature score and/or a low or negative PD-Li
status score,
the test report may indicate that the subject has a score that is associated
with response or
better response to treatment with a pembrolizumab, pembrolizumab variant
and/or antigen-
binding fragment thereof / talimogene laherparepvec combination therapy, while
if the CD8+
T-cell density score and/or IFN-y gene signature scores are above the
threshold, and/or PD-
Li status score is high. then the test report indicates that the patient has a
score that is
associated with no response or poor response to treatment with a
pembrolizumab,
pembrolizumab variant and/or antigen-binding fragment thereof / talimogene
laherparepvec
combination therapy.
59

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0229] In another embodiment, the tested tumor sample is from a cancer that is
responsive to
monotherapy with a checkpoint inbibitor (e.g., with anti-PD-Li therapy or anti-
PD-1 therapy
(e.g., pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)),
which may optionally be associated with one or more of a CD8+ T-cell
infiltration density of
fewer than about 1500, about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells per 1 mm2 sample (or
per 1 mL
sample), a low or negative PD-L1 status and a negative IFNy gene signature,
including, but
not limited to, melanoma (e.g., cutaneous, metastatic, uveal), lung cancer
(e.g., non-small cell
lung cancer, small cell lung cancer), head and neck cancer (e.g., recurrent or
metastatic
squamous cell carcinoma of the head and neck, nasopharyngeal cancer, thyroid
cancer,
salivary cancer, esophageal cancer), breast cancer (e.g., ER+/HER2- breast
cancer, triple-
negative breast cancer), ovarian cancer, cervical cancer, bladder cancer
(e.g., urothelial
cancer), renal cell cancer, gastrointestinal cancer (e.g., hepatocellular
cancer, colorectal
cancer, anal cancer), biliary tract cancer, multiple myeloma, lymphoma (e.g.,
mediastinal
large B-cell lymphoma, Hodgkin's lymphoma) or mesothelioma.
[0230] In some embodiments, the test report is a written document prepared by
the diagnostic
laboratory and sent to the patient or the patient's physician as a hard copy
or via electronic
mail. In other embodiments, the test report is generated by a computer program
and
displayed on a video monitor in the physician's office. The test report may
also comprise an
oral transmission of the test results directly to the patient or the patient's
physician or an
authorized employee in the physician's office. Similarly, the test report may
comprise a
record of the test results that the physician makes in the patient's file.
[0231] Detecting the presence or absence of a marker of the invention may be
performed
using a kit that has been specially designed for this purpose. In one
embodiment, the kit
comprises a set of oligonucleotide probes capable of hybridizing to the target
transcripts in
the gene signature. The kit may further comprise oligonucleotide probes
capable of detecting
transcripts of other genes, such as control genes, or genes used for
normalization purposes.
The set of oligonucleotide probes may comprise an ordered array of
oligonucleotides on a
solid surface, such as a microchip, silica beads (such as BeadAn-ay technology
from Illumina,
San Diego, Calif.), or a glass slide (see, e.g., WO 98/20020 and WO 98/20019).
In some
embodiments, the oligonucleotide probes are provided in one or more
compositions in liquid
or dried form.

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Immu no hi stoc he m istr y (IHC)
[0232] An IHC assay typically begins with antigen retrieval, which may vary in
terms of
reagents and methods. The antigen retrieval process may involve pressure
cooking, protease
treatment, microwaving, or heating histologic sections in baths of appropriate
buffers, with
the standard goal of unmasking antigens hidden by formalin crosslinks or other
fixation.
(See, e.g., Leong et al. Appl. Immnunohistochem. 4(3):201 (1996).)
[0233] Two general methods of IHC may be used: direct and indirect assays. In
a direct MC
assay, binding of antibody to the target antigen is determined directly. This
direct assay uses
a labeled reagent, such as a fluorescent tag or an enzyme-labeled primary
antibody, which
can be visualized without further antibody interaction. In a typical indirect
assay,
unconjugated primary antibody binds to the antigen and then a labeled
secondary antibody
binds to the primary antibody. Where the secondary antibody is conjugated to
an enzymatic
label, a chromagenic or fluorogenic substrate is added to provide
visualization of the antigen.
Signal amplification occurs because several secondary antibodies may react
with different
epitopes on the primary antibody.
[0234] The primary and/or secondary antibody used for immunohistochemistry
typically will
be labeled with a detectable moiety. Numerous labels are available which can
be generally
grouped into the following categories: (a) Radioisotopes, such as 35S, 14C,
1311, 3H, and 1231
(The antibody can be labeled with the radioisotope using the techniques
described in Current
Protocols in Immunology, Volumes 1 and 2, Coligen et al, Ed. Wiley-
lnterscience, New
York, N.Y., Pubs. (1991) for example and radioactivity can be measured using
scintillation
counting. Other radionuclides include 99Tc, 90Y, 32p, 13N, 18F, 51Cr, 57To,
225Ra, 60Co, 59Fe,
57se, 152Eu, 67cu, 217ci, 211At, 212pb, 47sc, 109pd, 234Th, 40K, 157Gd, 55mh,
52Tt, 82Rb, 201Th,
92Sr, 67Ga, 1921r, 166HO, 10B, 99mirc, 42K, 'Re, 188Re, 75se, 24Na, nc, 13N,
150, 57c0, 670a,
177Lu and 56Fe.); Colloidal gold particles: and fluorescent or
chemiluminescent labels
including, but not limited to, rare earth chelates (europium chelates),
fluorescein and its
derivatives, rhodamine and its derivatives, isothiocyanate, phycoerythrin,
phycocyanin,
allophycocyanin, o-phthaladehyde, fluorescamine, dansyl, umbelliferone,
luciferin, luminal
label, isoluminal label, an aromatic acridinium ester label, an imidazole
label, an acridimium
salt label, an oxalate ester label, an aequorin label, 2,3-
dihydrophthalazinediones, Texas Red,
dansyl, Lissamine, umbelliferone, phycocrytherin, phycocyanin, or commercially
available
flu orophores such SPECTRUM ORANGE and SPECTRUM GREEN and/or derivatives
of any one or more of the above. The fluorescent labels can be conjugated to
the antibody
61

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
using the techniques disclosed in Current Protocols in Immunology, Supra, for
example.
Fluorescence can be quantified using a fluorimeter.
[0235] Various enzyme-substrate labels are available (See U.S. Pat. No.
4,275,149). The
enzyme generally catalyzes a chemical alteration of the chromogenic substrate
that can be
measured using various techniques. For example, the enzyme may catalyze a
color change in
a substrate, which can be measured spectrophotometrically. Alternatively, the
enzyme may
alter the fluorescence or chemiluminescence of the substrate. Techniques for
quantifying a
change in fluorescence are described above. The chemiluminescent substrate
becomes
electronically excited by a chemical reaction and may then emit light which
can be measured
(using a chemiluminometer, for example) or donates energy to a fluorescent
acceptor.
[0236] Examples of enzymatic labels include luciferases (e.g., firefly
luciferase and bacterial
luciferase; U.S. Pat. No. 4,737,456), luciferin, 2,3-dihydrophthalazinediones,
malate
dehydrogenase, urease, peroxidase such as horseradish peroxidase (HRPO),
alkaline
phosphatase, 15-galactosidase, glucoarnylase, lysozyme, saccharide oxidases
(e.g., glucose
oxidase, galactose oxidase, and glucose-6-phosphate dehydrogenase),
heterocyclic oxidases
(such as uricase and xanthine oxidase), lactoperoxidase, microperoxidase, and
the like.
Techniques for conjugating enzymes to antibodies are described in O'Sullivan
et al, Methods
for the Preparation of Enzyme- Antibody Conjugates for use in Enzyme
Immunoassay, in
Methods in Enzym. (ed J. Langbne & H. Van Vunakis), Academic press, New York,
73:147-
166 (1981).
[0237] Numerous enzyme-substrate combinations are available to those skilled
in the art.
For a general review of these, see U.S. Pat. Nos. 4.275,149 and 4,318,980.
Examples of
enzyme- substrate combinations are: (i) Horseradish peroxidase (HRP) with
hydrogen
peroxidase as a substrate, wherein the hydrogen peroxidase oxidizes a dye
precursor, such as,
e.g., 3,3' diamino benzidine (DAB), which produces a brown end product; 3-
amino-9-
ethylcarbazole (AEC), which upon oxidation forms a rose-red end product; 4-
chloro-l-
napthol (CN), which precipitates as a blue end product; and p-
Phenylenediamine
dihydrochloride/pyrocatecol, which generates a blue-black product;
orthophenylene diamine
(OPD) and 3.3',5,5'-tetramethyl benzidine hydrochloride (TMB); (ii) alkaline
phosphatase
(AP) and para-Nitrophenyl phosphate, naphthol AS-MX phosphate, Fast Red TR and
Fast
Blue BB, napthol AS-BI phosphate, napthol AS-TR phosphate, 5-bromo- 4-chloro-3-
indoxyl
phosphate (BCIP), Fast Red LB, Fast Garnet GBC, Nitro Blue Tetrazolium (NBT).
and
iodonitrotetrazolium violet (INT); and (iii) 13-D-ga1actosidase (13-D-Gal)
with a clutomogenic
62

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
substrate (e.g., p-nitrophenyl-P-D- galactosidase) or fluorogenic substrate
(e.g., 4-
methylumbelliferyl-P-D-galactosid ase).
[0238] Any method known in the art for conjugating the antibody molecules to
the various
moieties may be employed, including those methods described by Hunter, et al,
(1962)
Nature 144:945; David, et al, (1974) Biochemistry 13: 1014; Pain, et al,
(1981) J. Immunol.
Meth. 40:219; and Nygren, J., (1982) Histochem. and Cytochem. 30:407.
[0239] In some embodiments, the label is indirectly conjugated with the
antibody. The
skilled artisan will be aware of various techniques for achieving this. For
example, the
antibody can be conjugated with biotin and any of the four broad categories of
labels
mentioned above can be conjugated with avidin, or vice versa. Biotin binds
selectively to
avidin and thus, the label can be conjugated with the antibody in this
indirect manner.
Alternatively, to achieve indirect conjugation of the label with the antibody,
the antibody is
conjugated with a small hapten and one of the different types of labels
mentioned above is
conjugated with an anti-hapten antibody. Thus, indirect conjugation of the
label with the
antibody can be achieved.
[0240] After antigen retrieval and an optional blocking step, the tissue
section is exposed to
pembrolizumab, a pembrolizumab variant and/or an antigen-binding fragment
thereof as the
primary antibody for a sufficient period of time and under suitable conditions
to allow the
primary antibody to bind to the PD-L1 protein in the tissue section.
Appropriate conditions
for achieving this can be determined by routine experimentation. The slide is
then washed to
remove unbound and excess amounts of the primary antibody.
[0241] In some embodiments, the primary antibody is linked to a detectable
label, such as
paramagnetic ions, radioactive isotopes, fluorochromes, and the like, and the
slide is
evaluated for PD-Li staining using the appropriate imaging apparatus.
[0242] In other embodiments, immune complexes between PD-L1 and the primary
antibody
may be detected using a second binding agent that is linked to a detectable
label. The second
binding agent is preferably a secondary antibody, which is applied to the
slide at a
concentration and for a period of time sufficient to allow the formation of
secondary immune
complexes. The slide is then typically washed to remove any non-specifically
bound
secondary antibody, and the label in the secondary immune complexes is
detected.
[0243] The secondary antibody may be labeled using avidin, streptavidin or
biotin, which is
independently labeled with a detectable moiety, such as a fluorescent dye
(stain), a
luminescent dye or a non-fluorescent dye. In principle, any enzyme that can be
conjugated to
63

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
or can bind indirectly to the secondary antibody (e.g., via conjugated avidin,
streptavidin,
biotin) could be used. The enzyme employed can be, for example, alkaline
phosphatase (AP),
horseradish peroxidase (HRP), beta-galactosidase and/or glucose coddase. The
enzyme can
also be directed at catalyzing a luminescence reaction of a substrate, such
as, but not limited
to, luciferase and aequorin, having a substantially non-soluble reaction
product capable of
luminescing or of directing a second reaction of a second substrate, such as
but not limited to,
luciferin and ATP or coelenterazine and Ca2+, having a luminescing product.
Finally, a
detection reagent is applied that includes a chromagen or a fluorescently
tagged molecule to
visualize the localization of the immune complexes.
[0244] The IHC assay may be performed using an automated pathology system,
which may
include automated staining (conventional stains, histochemical techniques,
immunostainers);
automated in situ hybridization systems; automatic slide preparation
(covenlip, slide drying)
and integrated slide and cassette labeling, as described in Roja et al.,
Review of imaging
solutions for integrated, quantitative immunohistochemistry in the Pathology
daily practice,
Folia Histochemica et Cytobiologica, Vol. 47, No. 3, 349-354, 2009.
[0245] in certain exemplary embodiments, an IHC assay employs the commercially
available
Dako EnVisionTm FLEX detection system, which is intended for use together with
a Dako
Autostainer instrument (Dako, an Agilent Technologies Company, Glostrup,
Denmark).
These reagents can be used off the shelf for other autostainers or for
manually-performed
staining (not performed with an autostainer).
Sample Collection and Preparation of Tissue Sections
[0246] A tumor tissue sample used to prepare stained tissue sections for
scoring PD-L1
expression can be collected from a subject before and/or after exposure of the
subject to one
or more therapeutic agents, e.g. a pembrolizumab, pembrolizumab variant and/or
antigen-
binding fragment thereof, talimogene laherparepvec and/or a chemotherapeutic
agent.
Accordingly, tumor samples may be collected from a subject over a period of
time. The
tumor sample can be obtained by a variety of procedures including, but not
limited to,
surgical excision, aspiration or biopsy. The tissue sample may be sectioned
and examined for
PD-Li as a fresh specimen. In other embodiments, the tissue sample is frozen
for further
sectioning. In other embodiments, the tissue sample is preserved by fixing and
embedding in
paraffin or the like.
[0247] The tissue sample may be fixed by conventional methodology, with the
length of
fixation depending on the size of the tissue sample and the fixative used.
Neutral buffered
64

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
formalin, glutaraldehyde, Bouin's or paraformaldehyde are non-limiting
examples of
fixatives. In preferred embodiments, the tissue sample is fixed with formalin.
In some
embodiments, the fixed tissue sample is also embedded in paraffin to prepare a
formalin-
fixed and paraffin- embedded (FFPE) tissue sample. Examples of paraffin
include, but are
not limited to, Paraplast, Broloid and Tissuemay.
[0248] Typically, the tissue sample is fixed and dehydrated through an
ascending series of
alcohols, infiltrated and embedded with paraffin or other sectioning media so
that the tissue
sample may be sectioned. Alternatively, the tumor tissue sample is first
sectioned and then
the individual sections are fixed.
[0249] in some embodiments, the scoring process of the invention is performed
on FFPE
tissue sections of about 3-4 millimeters, and preferably 4 micrometers, which
are mounted
and dried on a microscope slide.
Pembrolizumab and Pembrolizumab Variant Antibodies
[0250] The primary antibody for an IHC assay described herein is
pembrolizumab, a
pembrolizumab variant and/or an antigen-binding fragment thereof, which binds
to the
mature form of PD-Li (lacking the pre-secretory leader sequence, also referred
to as leader
peptide) that is expressed on the surface of certain mammalian cells. The
terms "PD-Li" and
"mature PD-Ll" are used interchangeably herein, and shall be understood to
mean the same
molecule unless otherwise indicated or readily apparent from the context. As
used herein, an
anti-human PD-Li antibody or an anti-hPD-L1 antibody refers to an antibody
that
specifically binds to mature human PD-L1, e.g., pembrolizumab, a pembrolizumab
variant
and/or an antigen-binding fragment thereof. A mature human PD-Li molecule
consists of
amino acids 19-290 of the following sequence:
MRIFA VFIFMT YW HLLN AFTVTVPKDLY VVE YGS NMTIEC KFPVEKQLDLA A LI VY
WEMEDICNIIQFVHGEEDLKVQHSSYRQRARLLICDQLSLGNAALQITDVICLQDAGVY
RCMISYGGADYKRITVKVNAPYNKINQRILVVDPVTSEHELTCQAEGYPKAEVIWTS
SDHQVLSGKTT'TTNSKREEKLFNVTSTLRINTTTNEIFYCTFRRLDPEENHTAELVIPE
LPLAHPPNERTHLVILGAILLCLGVALTFIFRLRKGRMMDVKKCGIQDTNSKKQSDTH
LEET (SEQ ID NO:11).
[0251] An antibody that "specifically binds to human PD-Li ," or an antibody
that
"specifically binds to a polypeptide comprising the amino acid sequence of
human PD-Li ," is
an antibody that exhibits preferential binding to human PD-L1 as compared to
other antigens,
but this specificity does not require absolute binding specificity. An anti-
hPD-L1 antibody is

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
considered "specific" for human PD-L1 if its binding is determinative of the
presence of
human PD-Li in a sample, e.g. without producing undesired results such as
false positives in
an 1HC diagnostic assay. Antibodies, or binding fragments thereof, useful as a
primary
antibody in the processes and methods of the present invention will bind to
human PD-Ll
with an affinity that is at least two fold greater, preferably at least ten
times greater, more
preferably at least 20-times greater, and most preferably at least 100-times
greater than the
affinity with any non-PD-Ll protein. As used herein, an antibody is said to
bind specifically
to a polypeptide comprising a given sequence, e.g. mature human PD-Li. if it
binds to
polypeptides comprising that sequence but does not bind to proteins lacking
that sequence.
Tissue sections of tumor samples from human subjects may be scored for PD-L1
expression
using pembrolizumab, a pembrolizumab variant and/or an antigen-binding
fragment thereof.
Diagnostic Testing for PD-Ll Expression
[0252] In one embodiment, the tested tumor sample is from a cancer that is
associated with a
low or negative PD-L1 status, and optionally a CD8+ T-cell infiltration
density of fewer than
about 1500, about 1400, about 1300, about 1200, about 1100, about 1000, about
900, about
800, about 700, about 600, or about 500 cells per 1 mm2 sample (or per 1 mL
sample) and/or
a low or negative PD-Li status, and is from melanoma (e.g., cutaneous,
metastatic, uveal),
non-small cell lung cancer, head and neck cancer (e.g., recurrent or
metastatic squamous cell
carcinoma of the head and neck), colorectal cancer, breast cancer (e.g.,
HER2+, HER2-
(HR+), triple-negative), ovarian cancer, bladder cancer (e.g., transitional
cell cancer,
urothelial cancer), prostate cancer (e.g., castration-resistant), sarcoma
(e.g., soft tissue, bone),
renal cell cancer, gastric cancer, esophageal cancer, anal canal cancer,
biliary tract cancer or
pancreatic cancer.
[0253] In another embodiment, the tested tumor sample is responsive to
monotherapy with a
checkpoint inbibitor (e.g., with anti-PD-L1 therapy or anti-PD-1 therapy
(e.g.,
pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)), and may
optionally be associated with one or more of a CD8+ T-cell infiltration
density of fewer than
about 1500, about 1400, about 1300, about 1200, about 1100, about 1000. about
900, about
800, about 700, about 600, or about 500 cells per 1 mm2 sample (or per 1 mL
sample), a low
or negative PD-L1 status and a negative IFNy gene signature, including, but
not limited to,
melanoma (e.g., cutaneous, metastatic, uveal), lung cancer (e.g., non-small
cell lung cancer,
small cell lung cancer), head and neck cancer (e.g., recurrent or metastatic
squamous cell
66

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
carcinoma of the head and neck, nasopharyngeal cancer, thyroid cancer,
salivary cancer,
esophageal cancer), breast cancer (e.g., ER+/HER2- breast cancer, triple-
negative breast
cancer), ovarian cancer, cervical cancer, bladder cancer (e.g., urothelial
cancer), renal cell
cancer, gastrointestinal cancer (e.g., hepatocellular cancer, colorectal
cancer, anal cancer),
biliary tract cancer, multiple myeloma, lymphoma (e.g., mediastinal large B-
cell lymphoma,
Hodgkin's lymphoma) or mesothelioma.
[0254] Each of the steps of obtaining a tissue sample, preparing one or more
tissue sections
therefrom for IHC assay, performing the IHC staining, and scoring the results
may be
performed by separate individuals/entities at the same or separate locations.
For example, a
surgeon may obtain by biopsy a tissue sample from a cancer patient's tumor and
then send
the tissue sample to a pathology lab, which may fix the tissue sample and then
prepare one or
more slides, each with a single tissue section, for IHC assay. The slide(s)
may be analyzed
by IHC soon after preparation, or stored for future assay. The lab that
prepared a tissue
section for IHC assay may conduct the assay or send the slide(s) to a
different lab to conduct
the assay. A pathologist or trained professional who scores the stained
slide(s) for PD-Li
staining may work for the diagnostic lab, or may be an independent contractor.
Alternatively,
a single diagnostic lab obtains the tissue sample from the subject's physician
or surgeon and
then performs all of the steps involved in preparing tissue sections, staining
the slide(s) and
scoring PD-Li expression in the stained tissue section(s) or sending the
stained slide(s) to a
trained professional for PD-Li scoring.
[0255] In some embodiments, the individuals involved with preparing and
analyzing the
tissue section by IHC assay do not know the identity of the subject whose
sample is being
tested, i.e., the sample received by the laboratory is made anonymous in some
manner before
being sent to the laboratory. For example, the sample may be merely identified
by a number
or some other code (a "sample ID") and the results of the IHC assay is
reported to the party
ordering the test using the sample ID. In preferred embodiments, the link
between the
identity of a subject and the subject's tissue sample is known only to the
individual or to the
individual's physician.
[0256] In some embodiments, after the test results have been obtained, the
diagnostic
laboratory generates a test report, which may include a result that the tissue
sample was
positive or negative for PD-Li expression. The test report may also include
guidance on how
to interpret the results for predicting if a subject is likely to respond to a
pembrolizumab,
pembrolizumab variant and/or antigen-binding fragment thereof / talimogene
laherparepvec
67

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
combination therapy. For example, in one embodiment, if the patient's tumor is
below a pre-
specified threshold, the test report may indicate that the patient has a PD-L1
expression score
that is correlated with response or better response to treatment with
pembrolizumab,
pembrolizumab variant and/or antigen-binding fragment thereof / talimogene
laherparepvec
combination therapy, while if the PD-Li expression score is at or above a pre-
specified
threshold, the PD-Li expression score is correlated with no response or poor
response to
treatment with pembrolizumab, pembrolizumab variant and/or antigen-binding
fragment
thereof / talimogene laherparepvec combination therapy.
[0257] In some embodiments, the test report is a written document prepared by
the diagnostic
laboratory and sent to the patient or the patient's physician as a hard copy
or via electronic
mail. In other embodiments, the test report is generated by a computer program
and
displayed on a video monitor in the physician's office. The test report may
also comprise an
oral transmission of the test results directly to the patient or the patient's
physician or an
authorized employee in the physician's office. Similarly, the test report may
comprise a
record of the test results that the physician makes in the patient's file.
Methods to Quantify Lymphocytes
Flow Cytometry
[0258] Lymphocyte subsets are typically measured by immuno-fluorescent
labeling of cells
with fluorochromes conjugated to specific monoclonal antibodies and
quantifying the
proportion of specifically labeled cells by flow cytometry. Manual
alternatives to flow
cytometry are also available to quantify CD4 cells. They are simple light or
fluorescence
microscopy methods that just require cell counting.
[0259] In flow cytometry, specific monoclonal antibodies made against the
specific CD
antigens present on the cells are labeled with fluorescent dyes. The labeled
monoclonal
antibodies are allowed to react with the mononuclear cells (lymphocytes and
monoc)'tes), and
the cells that react can be classified by the flow cytometer into
subpopulations depending on
which antibodies are bound. Flow cytometry generally gives the percentage of
CD4+ or
CD8+ cells. To obtain absolute cell counts. dual and single platform
technologies are used.
A dual platform technology employs a flow cytometer and a hematology analyzer.
CD4
absolute count using dual platform approach is a product of three
measurements: the white
blood cell count, the percentage of white blood cells counts that are
lymphocytes
(differential), and the percentage of lymphocytes that are CD4 cells
(determined by flow
68

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
cytometry). If a single platform technology is used, absolute counts of
lymphocyte subsets
are measured in a single tube by a single instrument. Usually it is
accomplished by spiking a
fixed volume of sample with a known number of fluorescent beads (bead-based
systems) or
by precisely recording the volume of the sample analyzed. Recent
recommendations suggest
that single platform technology should be the gold standard for the CD4
absolute count.
[0260] Several varieties of flow cytometers are available, with the
FACSCalibur (Becton
Dickinson) and EPICS XL (Beckman Coulter) being the most popular. These
instruments
offer high sample throughput, workflow management through automation, and
simple
software applications. Both instruments can detect four colors and measure
relative cell size
and cellular complexity. The systems are designed to use whole blood,
collected in liquid
EDTA. Besides using the traditional flow cytometers (open platforms that can
employ dual
or single platform technology), the simplified dedicated platforms are
developed for CD4+ T-
cell counts. The commercially available dedicated platforms include FACScount
(Becton
Dickinson), CyFLow Counter (Partec), and Guava Auto CD4/CD8%
(Millipore/Merck). The
dedicated platforms allow CD4+ T-cell counting with reduced technical
complexity,
producing absolute CD4 counts and a CD4/CD8 ratio without requiring an
external computer.
The system uses whole blood, eliminates the need for lysis and wash steps, and
has a unique
software algorithm that automatically identifies the lymphocyte populations of
interests.
Manual Methods to Quantify Lymphocytes
[0261] Manual alternatives to flow cytometry available on the market are: the
Cyto-Spheres
(Coulter Corporation. USA) and the Dynabeads (Dynal AS, Norway). The Dynal T4
kit (the
Dynabeads) is used to manually count CD4 cells in a cell counting chamber
under a
microscope. This method measures CD4 absolute count; no lymphocyte percentages
can be
determined. It requires an epifluorescent microscope (recommended), although
it can be
performed with only a light microscope; a hemocytometer, a vortex, a tube
rocker, a timer,
and a magnet. Magnetic beads are coated with monoclonal antibodies as a solid
phase to
isolate CD4 and CD8 cells from whole blood, whereas CD4-positive monocytes are
pre-
depleted using CD14 magnetic beads. After isolation of CD4 cells, the cells
are lysed,
stained, and counted. Blood samples should be fresh, preferably not older than
24 hours.
The Coulter Manual CD4 Count Kit (for the Cyto-Spheres method) requires a
light
microscope, timer, and a hemocytometer and measures CD4 absolute counts (no
percentages)
from whole blood collected in EDTA tubes. Antibody-coated latex particles are
used to bind
CD4 cells resulting in a "rosette" of latex beads around each CD4 cell; the
rosette is readily
69

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
recognized by light microscopy. A monocyte blocking reagent minimizes the
interference
from monocytes that contain CD4 antigens because they can be recognized during
CD4 cell
counting.
[0262] It will be readily apparent to those skilled in the art that other
suitable modifications
and adaptations of the methods described herein may be made using suitable
equivalents
without departing from the scope of the embodiments disclosed herein. Having
now
described certain embodiments in detail, the same will be more clearly
understood by
reference to the following examples, which are included for purposes of
illustration only and
are not intended to be limiting.
Diagnostic Testing for CD8+ Expression
[0263] In one embodiment, the tested tumor sample is from a cancer associated
with a CD8+
T-cell infiltration density of fewer than about 1500, about 1400. about 1300,
about 1200,
about 1100, about 1000, about 900, about 800, about 700, about 600, or about
500 cells per 1
mm2 sample (or per 1 mL sample), optionally having a low or negative PD-L1
status and/or a
negative IFNy gene signature, including, but not limited to, melanoma (e.g.,
cutaneous,
metastatic, uveal), non-small cell lung cancer, head and neck cancer (e.g.,
recurrent or
metastatic squamous cell carcinoma of the head and neck), colorectal cancer,
breast cancer
(e.g., HER2+, HER2- (HR+), triple-negative), ovarian cancer, bladder cancer
(e.g.,
transitional cell cancer, urothelial cancer), prostate cancer (e.g.,
castration-resistant), sarcoma
(e.g., soft tissue, bone), renal cell cancer, gastric cancer, esophageal
cancer, anal canal cancer,
biliary tract cancer or pancreatic cancer.
[0264] In one embodiment, the tested tumor sample is from a cancer that is
responsive to
monotherapy with a checkpoint inhibitor (e.g., with anti-PD-Li therapy or anti-
PD-1 therapy
(e.g., pembrolizumab, a pembrolizumab variant or an antigen-binding fragment
thereof)),
which may optionally be associated with one or more of a CD8+ T-cell
infiltration density of
fewer than about 1500. about 1400, about 1300, about 1200, about 1100, about
1000, about
900, about 800, about 700, about 600, or about 500 cells per 1 mm2 sample (or
per 1 mL
sample), a low or negative PD-Li status and a negative IFNy gene signature,
including, but
not limited to, melanoma (e.g., cutaneous, metastatic, uveal), lung cancer
(e.g., non-small cell
lung cancer, small cell lung cancer), head and neck cancer (e.g., recurrent or
metastatic
squamous cell carcinoma of the head and neck, nasopharyngeal cancer, thyroid
cancer,
salivary cancer, esophageal cancer), breast cancer (e.g., ER+/HER2- breast
cancer, triple-
negative breast cancer), ovarian cancer, cervical cancer, bladder cancer
(e.g., urothelial

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
cancer), renal cell cancer, gastrointestinal cancer (e.g., hepatocellular
cancer, colorectal
cancer, anal cancer), biliary tract cancer, multiple myeloma, lymphoma (e.g.,
mediastinal
large B-cell lymphoma, Hodgkin's lymphoma) or mesothelioma.
[0265] Each of the steps of obtaining a sample, preparing the sample for flow
cytometry,
performing flow cytometry, and scoring the results may be performed by
separate
individuals/entities at the same or separate locations. For example, a surgeon
may obtain by
biopsy a sample from a cancer patient and then send the sample to a pathology
lab, which
may prepare the sample for flow cytometry. The slide(s) may be analyzed by IHC
soon after
preparation, or stored for future assay. The lab that prepared a sample for
flow cytometry
may conduct the assay or send the slide(s) to a different lab to conduct the
assay. A
pathologist or trained professional who performs flow cytometry may work for
the diagnostic
lab, or may be an independent contractor. Alternatively, a single diagnostic
lab obtains the
sample from the subject's physician or surgeon and then performs all of the
steps involved in
preparing the sample and scoring CD8+ in the sample or sending the sample to a
trained
professional for CD8+ scoring.
[0266] In some embodiments, the individuals involved with preparing and
analyzing the
sample by flow cytometry do not know the identity of the subject whose sample
is being
tested, i.e., the sample received by the laboratory is made anonymous in some
manner before
being sent to the laboratory. For example, the sample may be merely identified
by a number
or some other code (a "sample ID") and the results of the flow cytometry assay
are reported
to the party ordering the test using the sample ID. In preferred embodiments,
the link
between the identity of a subject and the subject's tissue sample is known
only to the
individual or to the individual's physician.
[0267] In some embodiments, after the test results have been obtained, the
diagnostic
laboratory generates a test report, which may include guidance on how to
interpret the results
for predicting if a subject is likely to respond to pembrolizumab,
pembrolizumab variant
and/or antigen-binding fragment thereof / talimogene laherparepvec combination
therapy.
For example, in one embodiment, if the patient's tumor is below a pre-
specified threshold,
the test report may indicate that the patient has a CD8+ expression score that
is correlated
with response or better response to treatment with pembrolizumab,
pembrolizumab variant
and/or antigen-binding fragment thereof / talimogene laherparepvec combination
therapy,
while if the CD8+ expression score is at or above a pre-specified threshold,
the CD8+
expression score is correlated with no response or poor response to treatment
with
71

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
pembrolizumab, pembrolizumab variant and/or antigen-binding fragment thereof /
talimogene laherparepvec combination therapy.
[0268] in some embodiments, the test report is a written document prepared by
the diagnostic
laboratory and sent to the patient or the patient's physician as a hard copy
or via electronic
mail. In other embodiments, the test report is generated by a computer program
and
displayed on a video monitor in the physician's office. The test report may
also comprise an
oral transmission of the test results directly to the patient or the patient's
physician or an
authorized employee in the physician's office. Similarly, the test report may
comprise a
record of the test results that the physician makes in the patient's file.
[0269] it will be readily apparent to those skilled in the art that other
suitable modifications
and adaptations of the methods described herein may be made using suitable
equivalents
without departing from the scope of the embodiments disclosed herein. Having
now
described certain embodiments in detail, the same will be more clearly
understood by
reference to the following examples, which are included for purposes of
illustration only and
are not intended to be limiting. All patents, patent applications and
references described
herein are incorporated by reference in their entireties for all purposes.
EXAMPLES
Example 1. Phase lb Clinical Trial Combining Talimogene Laherparepvec with
Pembrolizumab
[0270] A phase lb trial (MASTERKEY-265; ClinicalTrials.gov Identifier:
NCT02263508)
was designed in patients with advanced melanoma combining the intratumoral
injection of
talimogene laherparepvec with the systemic administration of the anti-PD-1
antibody
pembrolizumab, with baseline and repeated on-therapy biopsies, with the
primary objective
of testing the safety of this combination, and to explore its ability to boost
inflammatory
status of tumors.
[0271] Eligible patients (18 years) had histologically confirmed, surgically
unresectable,
stage IIIB¨IV cutaneous melanoma, measurable disease (>1 melanoma lesion with
longest
diameter >10 mm), and >1 injectable cutaneous, subcutaneous, or nodal melanoma
lesion(s)
that were >10 mm in longest diameter, either alone or in aggregate, for whom
surgery was
not recommended. Patients were required to have adequate performance status
and
hematologic, hepatic, renal, and coagulation function. Patients were excluded
if they had
uveal/mucosal melanoma; had been previously treated for advanced melanoma with
systemic
72

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
therapy, previously received talimogene laherparepvec or any prior systemic
anticancer
treatment for unresectable, stage IIIB¨IV melanoma; Eastern Cooperative
Oncology Group
(ECOG) performance status >2; active brain metastases; active herpetic skin
lesions; prior
complications from herpetic infection; or required systemic anti-herpetic
treatment other than
intermittent topical use. All patients provided written informed consent.
Study procedures
were approved by an institutional ethics committee at each site.
Study Design
[0272] The phase lb portion of the MASTERKEY-265 study was an open-label,
multicenter,
single-arm study that primarily evaluated the safety of intralesional
talimogene laherparepvec
in combination with intravenous pembrolizumab (Figure 6).
[0273] Briefly, to seroconvert herpes simplex virus-negative patients,
intralesional
talimogene laherparepvec 106 pfu/mL was administered on day 1 of study week 1.
Subsequent doses of talimogene laherparepvec 108 pfu/mL were administered on
day 1 of
weeks 4 and 6, and every 2 weeks thereafter. Up to 4 mL (total volume) of
talimogene
laherparepvec could be administered by intralesional injection at each
treatment visit. The
volume delivered to each injected lesion was contingent on the diameter of the
lesion
(Hoffner, B., Iodice, G.M., and Gasal, E. (2016). Administration and Handling
of Talimogene
Laherparepvec: An Intralesional Oncolytic Immunotherapy for Melanoma. Oncol
Nurs
Forum 43, 219-226). The injected volume per lesion ranged from 0.1 inL for
lesions <0.5 cm
to 4.0 mL for lesions > 5 cm in longest diameter. Talimogene laherparepvec
administration
continued until disappearance of injectable lesions, complete response (CR),
confirmed
disease progression (PD) per modified immune-related response criteria (irRC),
(Wolchok et
al., 2009) treatment intolerance, 24 months from the first dose of
pembrolizumab, or end of
study, whichever occurred first. If toxicity occurred, talimogene
laherparepvec doses could
be delayed for up to 4 weeks; delays >4 weeks resulted in permanent
discontinuation.
[0274] Pembrolizumab (200 mg) was administered intravenously every 2 weeks
beginning
on day 1 of week 6 (i.e., at the time of third dose of talimogene
laherparepvec).
Pembrolizumab treatment was to be continued until confirmed PD by irRC,
treatment
intolerance, 24 months from the first dose of pembrolizumab, or the end of
study, whichever
occurred first. Pembrolizumab could be withheld or discontinued per protocol-
specified rules
consistent with the US prescribing information (Kaufman, H.L., Kim, D.W.,
DeRaffele, G.,
Mitcham, J., Coffin. R.S., and Kim-Schulze, S. (2010). Local and distant
immunity induced
by intralesional vaccination with an oncolytic herpes virus encoding GM-CSF in
patients
73

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
with stage Inc and IV melanoma. Ann Surg Oncol 17, 718-730). If pembrolizumab
was
withheld >12 weeks, pembrolizumab treatment was permanently discontinued.
[0275] The primary endpoint was incidence of dose limiting toxicities (DLTs)
starting from
when both agents were given in combination. Incidence of DLTs in the first 6
DLT-
evaluable patients and additional safety data from all patients were evaluated
by a dose-level
review team. The combination would be declared tolerable if the incidence of
DLTs was
<33% during the DLT evaluation period. Secondary endpoints included confirmed
objective
response rate (ORR; the rate of CR plus partial response (PR)) as evaluated by
investigators
per irRC, (WoIchok et al., 2009) best overall response, and incidence of AEs.
[0276] DLTs were defined as any of the following treatment-related toxicities
occurring
during the 6-week period from the beginning of pembrolizumab treatment: grade
4 non-
hematologic toxicity; grade 3/4 pneumonitis; grade 3 non-hematologic toxicity
lasting >3
days despite optimal supportive care (except grade 3 fatigue); grade 3/4 non-
hematologic
laboratory value requiring medical intervention/hospitalization or persisting
>1 week; grade
3/4 febrile neutropenia; thrombocytopenia <25x109/L if associated with a life-
threatening
bleeding event or bleeding event requiring platelet infusion; any grade 5
toxicity; or any
toxicity requiring permanent discontinuation of talimogene laherparepvec or
pembrolizumab.
Study Clinical Assessments
[0277] Adverse events occurring from week 1 to 30 days after the last dose of
study
treatment were recorded and graded using Common Terminology Criteria for
Adverse Events
version 4Ø
[0278] Tumor response was evaluated per modified irRC (Wolchok, Hoos, A.,
O'Day,
S., Weber, J.S., Hamid, 0., Lebbe, C., Maio, M., Binder, M., Bohnsack, 0.,
Nichol, G., et al.
(2009). Guidelines for the evaluation of immune therapy activity in solid
tumors: immune-
related response criteria. Chin Cancer Res 15, 7412-7420) by investigators.
Complete
response was defined as the disappearance of all lesions; PR was defined as a
decrease in
tumor area > 50% relative to baseline; PD was defined as an increase in tumor
area >25%
relative to nadir; and SD was defined as any outcome not meeting the criteria
for response or
PD with >77 days elapsed after enrollment. Responses were confirmed within 4
weeks from
the date of first documentation of response. Tumor assessments were performed
at screening,
week 6 (prior to initiation of pembrolizumab), week 18, and every 12 weeks
thereafter.
Radiographic imaging for assessment of lesions was performed using computed
tomography,
74

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
positron emission tomography, magnetic resonance imaging, or ultrasound.
Clinical
measurement of cutaneous, subcutaneous, and palpable nodal tumor lesions was
conducted
with calipers. Initial measurement of PD was confirmed by assessment of
measureable/non-
measureable new lesions as well as index lesions >4 weeks later. If clinically
stable, patients
continued treatment while awaiting confirmation of PD.
Biomarker Analysis
Flow cytometry
[0279] T-cell subsets were analyzed by flow cytometry with the following
markers CD45.
CD3, CD4, CD8, and BD TruCOUNT. Additionally, checkpoint markers assessed
included
HLA-DR, PD-1, Tim3, BTLA, ICOS, 0X40, 41BB, and GITR on T-cell subsets as
defined
by expression of CD3, CD4, CD8, CCR7 and CD45RA.
RNA Profiling and IFNrGene Signature
[0280] Total RNA was isolated from 5 inn think formalin-fixed paraffin-
embedded (FFPE)
sections fixed on positively charged slides. Percentage tumor area was first
assessed and
either all tissue was scraped for isolation or if <50% tumor area was present,
tumor tissue was
macro-dissected for isolation. RNA isolation was performed using the High Pure
FFPET
RNA isolation kit from Roche Diagnostics (Indianapolis, IN). NanoString gene
expression
profiling was conducted using 50 ng of RNA run on the nCounter PanCancer
Immune
Profiling Panel from NanoString Technologies (Seattle, WA) per manufacturer's
instructions.
Normalized gene expression values from the NanoString assay were calculated by
subtracting
the log10 transformed raw counts for each gene from the 10g10 calculated mean
of the
housekeeping genes. An interferon gamma gene signature score was obtained
using a
calculation that compared the normalized value to a predefined weighted score
for each gene
within the signature.
Immunohistochemistry
[0281] PD-L1 expression in a tumor was assessed using IHC as described
previously (Daud,
A.I., Wolchok, J.D.. Robert, C., Hwu, W.J., Weber, J.S., Ribas, A., Hodi,
F.S.. Joshua, A.M.,
Kefford, R., Hersey, P., et al. (2016). Programmed Death-Ligand 1 Expression
and Response
to the Anti-Programmed Death 1 Antibody Pembrolizumab in Melanoma. J Clin
Oncol 34,
4102-4109) using an investigational version of the Dako PD-L1 22C3 assay
(Carpinteria,
CA).

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0282] For CD8 and granzyme B IHC analysis, a hematoxylin and eosin stain was
performed
and reviewed by a pathologist to verify the presence of melanoma and to define
tumor areas
as regions of interest for analysis. The anti-CD8 mouse monoclonal antibody
clone C8/144B
was used for CD8 IHC. The anti-granzyme B mouse monoclonal antibody clone GrB-
7
(Dako) was used for granzyme B IHC. Immunohistochemical detection was
performed with
a polymer-based detection method and a red chromogen. Slides were scanned
using a
ScanSc,ope CS or AT Turbo system (Aperio, Vista, CA, USA), the region of tumor
was
circled, and the density of positive cells (e.g., CD8-positive cells per mm2)
was evaluated by
automated image analysis.
Immunofluorescence
[0283] Available paired, pre- and post-treatment biopsies were evaluated using
MultiOmyxTM technology to stain 12 biomarkers using a single slide. Repeated
cycles of
staining using a pair of antibodies directly conjugated to either Cy3 or Cy5,
followed by
imaging and dye inactivation were performed according to published methods (Au
et at.,
2016; Gerdes et al., 2013).
Ouantification And Statistical Analysis
[0284] Using a 6+3 trial design, six to nine DLT-evaluable patients were
required to assess
the DLT profile of talimogene laherparepvec in combination with pembrolizumab,
assuming
a true DLT incidence rate between 11%-33%. Additional patients were enrolled
to evaluate
the association between bio markers and response.
[0285] The DLT analysis set included all DLT-evalu able patients enrolled in
phase lb who
had the opportunity to be on treatment >6 weeks from the initial dose of
pembrolizumab and
who received .>2 doses of talimogene laherparepvec and 2 doses of
pembrolizumab in
combination, or who experienced a DLT within 6 weeks of starting combination
therapy.
The safety analysis sets included all patients who received >1 dose talimogene
laherparepvec
or pembrolizumab. Predictive biomarker analyses included all patients with a
baseline
biomarker result; analyses of biomarker changes included all patients with a
baseline
biomarker result and >1 subsequent biomarker result.
[0286] Corresponding exact 95% confidence intervals (95% CI) for were
calculated for ORR
and disease control rate. PFS (time from enrollment to disease progression per
modified
irRC or death) and OS (time from enrollment to death) were estimated using the
Kaplan-
Meier method.
76

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0287] For cell density or H-score results from IHC, change from baseline was
assessed with
the sign-test of 1og2 ratio of post baseline over baseline (week 1) in
injected and non-injected
lesions separately. For flow cytometry results, change from baseline assessed
with a linear
mixed effects model with baseline as covariate for 10g10 ratio (absolute
counts or MESF) or
%-difference to/from baseline. For immunofluorescence-based mu ltiparameter
imaging,
effects on cell density were assessed with linear mixed effects models for
cube root of density
with factors visit and injection status (Ribas, A. (2015). Adaptive Immune
Resistance: How
Cancer Protects from Immune Attack Cancer Discov 5,915-919).
[0288] Association with the unconfirmed best response per investigator as of
August 2016
was evaluated with a logistic regression of response (CR or PR) vs. continuous
biomarker
results at either baseline or change from baseline at a given visit.
Transformed results were
used for analyses. Injected and non-injected lesions analyzed separately.
Kruskal-Wallis test
was also evaluated in cases of small sample size.
[0289] FOR controlled at 5% with the Benjamini-Hochberg procedure and flow
cytometry
analysis was stratified by a priority set of endpoints and reporting metric
(Abs, MESF, %).
Data and Software Availability
[0290] Statistical analyses were conducted using SAS version 9.2 (SAS
Institute, Cary, NC).
Biomarker statistical analyses were conducted using Matlab R2015a (The
Mathworks Inc.,
Natick, MA).
[0291] Example 2: Safety and Tolerability of Combining Talimogene
Laherparepvec
with Pembrolizumab
[0292] in the phase lb clinical trial, intratu moral talimogene laherparepvec
and intravenous
pembrolizumab were administered to 21 patients with advanced melanoma who had
skin
lesions amenable to injection. The combination was generally well tolerated,
with fatigue,
fevers and chills as most common adverse events. There were no dose-limiting
toxicities.
One patient had pneumonitis, a known toxicity of anti-PD-1 therapy. The
confirmed
objective response rate was 62%, with a complete response rate of 33%.
Patients who
responded to the combination of talimogene laherparepvec and pembrolizumab had
an
increase in CD8+ T-cells in tumors after talimogene laherparepvec. After
talimogene
laherparepvec treatment, PD-Lb increased on multiple immune cell subsets in
tumors along
with interferon gamma gene expression. Response to the combination therapy was
independent of the baseline infiltration by CD8+ T-cells or interferon gamma
signature.
77

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
These findings indicated that the intratumoral injection of talimogene
laherparepvec may
favorably change the tumor microenvironment by attracting T-cells into tumors,
thereby
facilitating the clinical activity of PD-1 blockade therapy.
[0293] The phase lb trial included a baseline biopsy before starting on
intratumoral
talimogene laherparepvec injections, with a first injection of up to 4 mL x
106 plaque-forming
units (pfu) per mL with the goal of inducing seroconversion and a protective
immune
response to the oncolytic viral vector, followed three weeks later with
repeated injections of
the full dose of up to 4 mL x 108 pfu / mL of talimogene laherparepvec every
two weeks
(Figure 1A). A second tumor biopsy was performed prior to administration of
the second full
dose of talimogene laherparepvec and before starting on pembrolizumab given at
200 mg
intravenously every 2 weeks coinciding with the next doses of talimogene
laherparepvec.
The run-in period with single agent talimogene laherparepvec administration
was designed to
analyze how intratu moral injection of talimogene laherparepvec alters the
tumor
microenvironment before starting on the combination therapy. A third tumor
biopsy was
planned during the combination therapy part of the study (Figures IA and 6).
The clinical
trial enrolled 21 patients with advanced melanoma and peripheral lesions
amenable to
intratumoral injection between December 2014 and March 2015 (see Table 2 for
full patient
characteristics). Patients were followed up for an average of 18.8 months
(range 17.7 to
20.7) at the time of reporting.
[0294] Table 2. Baseline Demographics and Clinical Characteristics
Talimogene laherparepvec
plus pembrolizumab
(N=21)
Sex, n (%)
Female 13(62)
Male 8 (38)
Median (range) age, y 58 (37-89)
ECOG performance status, n (%)
0 19(90)
1 2(1.0)
Disease substage, n (%)
111B 1 (5)
IlIC 6 (29)
I VM1a 2(10)
IVM1b 4(19)
IVM1c 8(38)
LDH, n (%)
<1J LN 16(76)
>ULN to 2x ULN 5(24)
>2x ULN
78

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
HSV serostatus, n (%)
Positive 16(76)
Negative 5 (24)
BRAF status, n(%)
Mutant 4(19)
Wild-type 17 (81)
PD-L1 status*, n (%)
Positive 17 (81)
Negative 2 (10)
Unknown 2(10)
ECOG, Eastern Cooperative Oncology Group; HSV, herpes simplex virus; LDH,
lactate
dehydrogenase; PD-L1, programmed death ligand 1; ULN, upper limit of normal.
* Cutoff for positivity was >1% PD-Li by immunohistochemistry.
[0295] With the combined therapy, there were no novel or dose limiting
toxicities in any of
the 21 patients. The most common toxicities were fatigue (62%), chills (48%),
and fever
(43%), which were anticipated with the intratumoral injection of talimogene
laherparepvec
(Andtbacka et al., 2015). Common pembrolizumab-related adverse events were
fatigue
(62%), rash (33%), and arthralgia (33%), which were anticipated with this
agent (Ribas et al.,
2016). The only serious adverse event attributed by the study investigators
potentially to the
combination was grade 1 cytokine release syndrome (one patient). Serious
adverse events
attributed to pembrolizumab included grade 3 autoimmune hepatitis, grade 3
aseptic
meningitis, and grade 4 pneumonitis (one patient each). In the patient with
treatment-related
aseptic meningitis, no herpes simplex virus was detected in the cerebrospinal
fluid, and the
patient had stopped therapy with talimogene laherparepvec and pembrolizumab
one month
earlier and had already switched therapy to dabrafenib and trametinib at the
time of first
presentation of this adverse event.
Example 3. Anti-tumor Activity with Combined Talimogene Laherparepvec and
Pembrolizumab
[0296] The confirmed objective response rate per immune-related response
criteria (irRC)
(Wolchok et al., 2009) was 61.9% (95% CI, 38.4%-81.9%), with a confirmed
complete
response rate of 33.3% (95% CI, 14.6%-57.0%) (Table 3). Responses occurred
across all
substages of melanoma (Figure 1B and C). Nine patients presented a transient
increase in
overall tumor size during the administration of talimogene laherparepvec, in
particular after
the first subtherapeu tic dose and before receiving the full dose of
talimogene laherparepvec
together with pembrolizumab, but sill these lesions responded later with the
combined
79

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
therapy (Figure 1D). Median progression free survival (PFS) and overall
survival (OS) were
not reached at the time of last follow up (Figure 1E and F). The combination
treatment
resulted in a greater than 50% size reduction in 82% of injected, 43.5% of non-
injected non-
visceral, and 33.4% of non-injected visceral lesions (Figure 6).
[0297] Table 3. Best Overall Response*
Talimogene laherparepvec plus pembrolizumab
,(N=21)
"rotaV Confirmed*
Patients with a response 15 13
Response rate, % (95% CI) 71 (48-89) 62 (38-82)
Best overall response, n (%)
Complete response 8 (38) 7 (33)
Partial response 7 (33) 6 (29)
Stable diseaset 1 (5) 3 (14)
Progressive disease 5 (24) 5 (24)
Disease control rate, n (%) 16 (76) 16 (76)
*Response was evaluated per immune-related response criteria by investigators.
tA best overall response of stable disease required an evaluation of stable
disease no earlier
than 77 days after enrollment.
*Responses were confirmed by a subsequent assessment at least 4 weeks later.
Example 4. Tumor Responses Independent of the Baseline CD8+ T-Cell
Infiltration,
PD-Li Status and Interferon Signature
[0298] PD-Li is induced by interferon gamma produced by tumor-infiltrating,
antigen-
specific T-cells, in what is termed adaptive immune resistance allowing cancer
cells to avoid
the cytotoxic activity of T-cells (Pardoll, 2012; Ribas, 2015). These T-cells
are then blocked
by PD-1 :PD-L1 interactions. Patients who respond to single agent PD-1
blockade therapy
have higher densities of baseline CD8+ T-cell infiltration, interferon gamma
gene expression
signatures and PD-Li expression (Herbst et at., 2014; Ribas et al., 2015;
Tumeh et al., 2014).
Vaseline biopsies of patients in the phase lb clinical trial described herein
were analyzed for
CD8+ T-cell density, PD-L1 positivity and interferon gamma gene signature. As
opposed to
the prior experience with single agent pembrolizumab therapy (Ribas et al.,
2015; Tumeh et
al., 2014), responses in the subject clinical trial were evident in patients
whose baseline
biopsies had very low CD8+ T-cell infiltrates or had a negative interferon
gamma gene
signature or has a negative PD-Li status. Among thirteen biopsies with a CD8+
density of

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
less than 1,000 cells/mm2, nine patients went on to respond to therapy and
four patients had
disease progression (Figure 2A). Out of the five baseline biopsies with a low
interferon
gamma signature, three patients went on to have a complete response and two
had disease
progression (Figure 2B). There was only one baseline biopsy that was scored as
being PD-Ll
negative, but that patient went onto have a complete response to the combined
therapy
(Figure 2C).
Example 5. Talimogene Laherparepvec Intratumoral Injections Increase CD8+ T-
cell
Infiltration in Patients Who Respond to Combined Therapy
[0299] As some patients whose baseline biopsies had relatively low CD8+ cell
density and
were not positive for an interferon gamma gene signature went onto have an
objective
response, the run-in period with single agent talimogene laherparepvec was
performed to
determine if talimogene laherparepvec had changed the tumor microenvironment
by bringing
in T-cells into metastatic melanoma lesions in patients who responded to
therapy. Indeed,
immunohistochemical (IHC) analysis comparing baseline biopsies with biopsies
performed
after talimogene laherparepvec alone showed an increase in the density of
infiltrating CD8+
T-cells in eight out of twelve injected lesions available for analysis, which
further increased
in several of the biopsies obtained at the time of combined therapy (Figure 3A
and B). In
three patients with a response to therapy, the CD8+ density went down in the
on-therapy
biopsy, and one additional patient had no change in CD8+ density. The three
patients
without a response all had a decrease in the CD8+ density in the on-therapy
biopsies.
Overall, the increase in CD8+ density was most evident in the injected lesions
of the patients
who went on to respond to therapy (Figure 3B), a relationship supported by
logistic
regression (p=0.0048, Figure 8A). The change in CD8+ infiltration density was
variable in
the non-injected lesions at week 6 even in patients who later responded to
therapy, with the
caveat that there were only three such biopsies available for interpretation
(Figure 3B). Some
biopsies were found to have low residual tumor content after treatment (as
indicated by open
symbols in Figure 3B and C), which, without intending to be bound by
scientific theory, may
have been due to either a complete pathological response at that site or a
biopsy that missed
the melanoma deposit. Four out of five responding biopsies with these tumor-
depleted
samples showed relatively high CD8+ T-cell density at week 6 as compared to
the biopsy of a
single patient with a progression included in this set. IHC was also performed
for the
cytotoxic granule component granzyme B (associated with a subset of CD8 T-
cells and NK-
81

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
cells), which has been shown to increase in tumors after PD-1 blockade (Tumeh
et al., 2014).
A trend suggesting increased granzyme B in tumors after talimogene
laherparepvec and
combination treatment was also observed in particular for the biopsies with
low residual
tumor content (Figure 3C). Furthermore, on analysis of tumor gene expression
data it was
determined that CD8+ alpha and interferon gamma mRNAs were elevated after
treatment,
providing additional supporting evidence for treatment-related change in the
tumor
microenvironment increasing the number of interferon gamma-producing cytotoxic
T-cells
(Figure 3D, 3E). CD8 alpha increased 1.7-fold (p=0.01) in injected lesions at
week 6
compared to baseline, and 1.44-fold (p=0.0012) in non-injected lesions.
Similarly, the
interferon gamma increases for injected and non-injected lesions were 1.63
(p=0.0004) and
1.41 (p=0.17), respectively.
Example 6. Characterization of Changes in Immune Cell Infiltrates in
Talimogene
Laherparepvec Injected and Non-Injected Lesions
[0300] To further characterize the changes in tumors, multiplexed
immunofluorescence
staining of a subset of paired biopsies at different time points from 13
patients was
performed. Broad changes in tumor inflammation were observed after talimogene
laherparepvec treatment at week 6, including increased infiltration by immune
cells and a
clear increase in cells expressing PD-Li in the eight out of ten of injected
tumors and in two
out of four non-injected tumors (Figure 4A). The immune infiltrate included
influx of a large
proportion of CD4+ and CD8+ T-cells, many co-expressing PD-1, as well as CD56
expressing cells and CD20 positive B-cells in the on treatment biopsies from
some patients.
Increases were also observed in the density of cells expressing the memory T-
cell marker
CD45R0 and in cells expressing the regulatory T-cell (Treg) marker Foxp3
(Figure 4A).
The magnitude of effector T-cell (Teff) increases, however, was much larger
relative to Treg,
resulting in an overall decrease in the Treg to Teff ratio in tumors after
talimogene
laherparepvec (Figure 9) consistent with previous reports (Kaufman et al.,
2010). The full set
of immunofluorescence analyses in biopsies is reported in Table 4, which
additionally shows
that there was no apparent increase in the density of macrophages based on
CD68 staining.
An example of increased CD8+ and PD-Li density (by immunofluorescence) at
weeks 6 and
30 relative to baseline is shown in Figure 4B. At weeks 6 and 30, tumor cells
co-staining for
S100 (blue) and PD-Li staining (red) are evident along with CD8 T-cells
(green) showing co-
expression of PD-Li. The biopsy taken during combined therapy of a responding
patient was
82

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
nearly completely infiltrated by CD8+ T-cells. Additional representative
images are
depicted in Figure 10.
Table 4. Cell subset changes within the tumor between injected lesions at week
6 and
baseline lesions (week 1).
Baseline
density Week 6 p-valuet
mean density ratio
Cell subset Icells/mmT to baselinet
CD3 204.21 2.57 2.28 x 10-23 __
CD4 68.96 2.73 7.56 x 1(121
CD8 126.15 2.47 3.70 x 1(129
FOXP3 _______________ 32.53 1.70 1.38 x 10-8
PD1 202.24 2.27 1.57 X HIP
PD-L1 172.28 2.73 1.99 x 1046
CD68 71.01 1.09 0.41
CD45R0 204.57 1.56 6.86 x 10.6
CD56 339.99 1.31 1.30 x 10-3
CD20 1.58 5.33 2.03 x 10-9
CTLA 22.49 0.77 0.09
CD3CD8 89.82 2.64 3.85 x 10-20
CD3CD8PD1 61.69 2.40 1.97 x i0-'5CD3CD4 68.96 2.73 7.56 x
let
CD3CD4PD1 32.57 2.76 9.26 x 10-29
CD3CD4FOX 13.00 1.90 1.46 x 10-7
P3
CD68PD_Ll 10.61 2.35 4.97 x 10-9
CD3+CD4+P 8.43 4.74 1.70 x10-2
D_Ll
CD3+CD4+C 29.72 2.02 4.53 x 104
D45R0
CD3+CD8+C 40.52 1.94 1.49 x 10-7
D45R0
CD3+CD8+P 9.43 4.19 1.37 x 10-16
D-Ll
CD3_CD8+C 1.01 3.34 2.53 x I0
D56
CD3+CD8+C 0.06 5.31 5.19 x10-5
D56+PD_Ll
CD3+CD4-1-P 32.57 2.76 9.26 x 10-20
D1
CD3+CD8+P 61.69 2.40 1.97 x 1045
Di
CD3-CD20+ 1.58 5.33 2.03 x 10-9
CD3- 0.16 6.35 1.15 x 10-7
CD2O+PD-L1
83

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Example 7. Changes in the Functional Phenotype of Circulating T-cells with
Combined
Therapy
[0301] Changes in immune cells were also analyzed in peripheral blood as a
potential
pharmacodynamic effect of the single agent and combined therapy. After
talimogene
laherparepvec single agent therapy, the majority of patients had an increase
in the number of
circulating CD8+ and CD4+ T-cells in peripheral blood, which did not increase
further when
adding pembrolizumab (Figure 5A and B). However, the addition of pembrolizumab
tended
to increase the number of dividing CD8 T-cells in circulation as indicated by
increase in
Ki67+CD3+CD8+ T-cells (Figure 5C). Analysis of the expression of different
immune
checkpoint receptors in the circulating CD3+ CD8+ T-cells revealed an increase
in PD-1 and
TIM-3 with single agent talimogene laherparepvec therapy (Figure 5D and E),
while there
was no change in BTLA (Figure 5F). No associations of response with baseline
cell levels or
changes over time passed our false discovery controls.
Example 8. Discussion
[0302] This first-in-human combination immunotherapy clinical trial
demonstrates a high
overall and complete response rate in patients with advanced melanoma, which
is mediated
by changes in tumor biopsies that are mechanistically correlated with the
hypothesis that the
injection of the oncolytic virus talimogene laherparepvec would change the
tumor
microenvironment by attracting T-cells that may induce a systemic response in
distant
metastases after subsequent blocking of PD-1 with pembrolizumab. Indeed,
during the run-in
period of the study with single agent talimogene laherparepvec intratumoral
administration
there was evidence of a systemic increase in circulating CD4 and CD8 T-cells
and increased
CD8 T-cell infiltration into tumors. These T-cells expressed PD-1 and the
tumor cells
expressed PD-L1, likely limiting the anti-tumor activity of single agent
talimogene
laherparepvec, which benefitted from blocking PD-1 resulting in an increased
clinical activity
beyond what would be expected with either therapy alone. The benefit of
increased
responses was achieved with a low rate of toxicities, most of which were
expected toxicities
with the use of single agent talimogene laherparepvec or pembrolizumab
(Andtbacka, R.H.,
Kaufman, H.L., Collichio, F., Amatruda, T., Senzer, N., Chesney, J., Delman,
K.A., Spitler,
L.E., Puzanov, I., Agarwala, S.S., et al. (2015). Talimogene Laherparepvec
Improves Durable
Response Rate in Patients With Advanced Melanoma. J Clin Oncol 33, 2780-2788;
Ribas, A.,
Hamid, 0., Daud, A., Hodi, F.S., Wolchok, J.D., Kefford, R., Joshua, A.M.,
Patnaik, A.,
84

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Hwu, W.J., Weber, J.S., et al. (2016). Association of Pembrolizumab With Tumor
Response
and Survival Among Patients With Advanced Melanoma. JAMA 315, 1600-1609).
[0303] PD-1 blockade therapy with pembrolizumab or nivolumab leads to an
objective
response of approximately 35-40% for treatment naive patients with metastatic
melanoma
without prior therapy (Ribas et al.. 2016; Robert, C., Long, G.V., Brady, B.,
Dutriaux, C.,
Maio, M., Mortier, L., Hassel, J.C., Rutkowski, P., McNeil, C., Kalinka-
Warzocha. E., et al.
(2015a). Nivolumab in previously untreated melanoma without BRAF mutation. N
Engl J
Med 372, 320-330; Robert, C., Schachter, J., Long, C.V., Arance, A., Grob,
J.J., Mortier, L.,
Daud, A., Carlino, M.S., McNeil, C., Lotem. M., et al. (2015b). Pembrolizumab
versus
1pilimumab in Advanced Melanoma. N Engl J Med 372, 2521-2532). Without
intending to
be bound by scientific theory, even considering that the need to select
patients that had
injectable lesions may have skewed the population of patients in this study,
an overall
response rate of 62% and a complete response rate of 33% is unlikely to be a
result of
administering anti-PD-1 therapy alone. In a study of 655 patients treated
with
pembrolizumab, there were 34 patients who had only skin and nodal metastases
(stage Mla),
and the overall response rate in this group of patients was 38% (Ribas et al.,
2016). A
randomized phase 3 clinical trial to further demonstrate that the combination
is more
effective than either single agent pembrolizumab or talimogene laherparepvec
is currently
ongoing comparing systemic administration of pembrolizumab with intralesional
injection of
talimogene laherparepvec or placebo (NCT 02263508).
[0304] Patients whose baseline biopsies had low densities of CD8+ T-cells,
lack of
significant interferon gamma expression, and low PD-L1 expression are unlikely
to respond
to single agent anti-PD-1 therapy (Ribas, A., Robert, C., Hodi, F.S., Wolchok,
J.D., Joshua,
A.M., Hwu, W.J., Weber, IS., Zarour, H.M., Kefford, R., Loboda, A., et al.
(2015).
Association of response to programmed death receptor 1 (PD-1) blockade with
pembrolizumab (MK-3475) with an interferon-inflammatory immune gene signature.
J Clin
Oncol 33, abstr 3001; Tumeh, P.C., Harview, C.L., Yearley, J.H., Shintaku,
I.P., Taylor, E.J.,
Robert, L., Chmielowski, B., Spasic, M., Henry, G., Ciobanu, V., et al.
(2014). PD-1
blockade induces responses by inhibiting adaptive immune resistance. Nature
515, 568-571).
Therefore, the combination therapy described herein should increase the
intratumoral
infiltration by CD8+ T-cells, which may attract enough T-cells with tumor
specificity that
may reverse the primary resistance to PD-1 blockade therapy (Chen, P.L., Roh,
W., Reuben,
A., Cooper, Z.A., Spencer, C.N., Prieto, P.A., Miller, J.P., Bassett, R.L.,
Gopalakrishnan, V.,

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Wani, K., et al. (2016). Analysis of Immune Signatures in Longitudinal Tumor
Samples
Yields Insight into Biomarkers of Response and Mechanisms of Resistance to
Immune
Checkpoint Blockade. Cancer Discov 6, 827-837; Ribas, A. (2015). Adaptive
Immune
Resistance: How Cancer Protects from Immune Attack. Cancer Discov 5, 915-919).
The data
presented herein demonstrate that talimogene laherparepvec can provide this
combinatorial
effect. In the series presented herein, the number of patients whose tumors
had low baseline
CD8+ density and low interferon gamma signature and still went onto have an
objective
response to the combined therapy was apparently higher compared to prior
experience with
single agent pembrolizumab (Ribas et al., 2015; Tumeh, P.C., Harview, C.L.,
Yearley, J.H.,
Shintaku, I.P., Taylor, E.j., Robert, L., Chmielowski, B., Spasic, M., Henry,
G., Ciobanu, V.,
et al. (2014). PD-1 blockade induces responses by inhibiting adaptive immune
resistance.
Nature 515, 568-571).
[0305] Evidence that local administration of talimogene laherparepvec was
contributing to a
systemic anti-tumor effect was provided by the increases in inflammation
observed in tumors
not injected with talimogene laherparepvec prior to introduction of
pembrolizumab.
Roughly, two out of the four week 6 non-injected lesions showed increased CD8+
density
and PD-Li (by immunofluorescence), and three out of five for increased
interferon gamma
mRNA.
[0306] The lack of requirement for baseline tumor infiltration will be further
evaluated in the
ongoing phase 3 study of the talimogene laherparepvec plus pembrolizumab
combination,
which is currently accruing 655 patients, half receiving the combination and
half
pembrolizumab with intratumoral placebo in the control arm
(ClinicalTrials.gov,
NCT02263508). Also, to further evaluate systemic effects of talimogene
laherparepvec, a
separate biomarker study is ongoing to evaluate baseline and post-talimogene
laherparepvec
uninjected tumors from over 100 patients (NCT02366195). This will enable
follow-up on
findings from the small set of tumor biopsies not injected with talimogene
laherparepvec in
the subject series, many of which showed increased tumor inflammation.
[0307] In conclusion, the high response rate in this phase 1 clinical trial
and the mechanistic
changes documented in patient biopsies indicates that the combination of
talimogene
laherparepvec and pembrolizumab can overcome some of the limitations of
pembrolizumab
or talimogene laherparepvec monotherapy and provide responses in patients
beyond what
would be expected with either talimogene laherparepvec or pembrolizumab
administered
alone.
86

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Example 9. Phase 3 Clinical Trial Combining Talimogene Laherparepvec with
Pembrolizumab
[0308] A phase 3 trial is being conducted in patients with previously
untreated, unresectable,
stage RIB to IVM1c melanoma to evaluate the efficacy of talimogene
laherparepvec with
pembrolizumab versus placebo with pembrolizumab, as assessed by Progression-
Free
Survival (PFS) (response evaluation by blinded independent central review
using modified
Response Evaluation Criteria in Solid Tumors 1.1 (RECIST)) and overall
survival (OS)
(Figure 12). Subjects are randomized 1:1 to receive the following: (1) Arm 1:
talimogene
laherparepvec plus pembrolizumab; and (2) Arm 2: placebo plus pembrolizumab.
[0309] The secondary objectives of the phase 3 trial include (1) evaluation of
the efficacy of
talimogene laherparepvec with pembrolizumab versus placebo with pembrolizumab
as
assessed by complete response rates, OS in subjects excluding Stage IVM1c,
overall response
rates, best overall response, durable response rate, and disease control rate;
(2) evaluation of
the safety of talimogene laherparepvec with pembrolizumab versus placebo with
pembrolizumab; and (3) evaluation of Patient Reported Outcomes (PRO) in phase
3 as
assessed by the European Organization for Research and Treatment of Cancer
(EORTC)
Quality of Life Questionnaire Core 30 (QLQ-C30) Global Health Status/Quality
of Life
(GHS/QoL) subscale.
[0310] Randomization is stratified by stage of disease: less advanced stages
(111B, BIC, and
IVM1a) versus more advanced stages (IVM1b and IVM1c) and by prior BRAF
inhibitor
therapy: no prior BRAF inhibitor versus BRAF inhibitor with or without MEK
inhibitor.
[0311] Key inclusion criteria include: Male or female age > 18 years with
histologically
confirmed diagnosis of melanoma and stage IIIB to IVM1c for whom surgery is
not
recommended. Subjects should have measurable disease and be a candidate for
intralesional
therapy administration into cutaneous, subcutaneous, or nodal lesions.
Subjects should have
Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, and
adequate
hematologic, hepatic, renal, and coagulation function. Subjects should be
treatment-naive
(should not have received any prior systemic anticancer treatment consisting
of
chemotherapy, immunotherapy or targeted therapy) given in a non-adjuvant
setting. Subjects
with BRAF mutation tumors may receive treatment with BRAF inhibitors either
alone or in
combination with MEK inhibitor as their only prior systemic therapy.
87

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0312] Key exclusion criteria include: No active cerebral metastases and or
carcinomatous
meningitis, subjects should not have uveal or mucosal melanoma. Subjects
should have no
history of immunodeficiency states. Subjects should not have received prior
treatment with
talimogene laherparepvec, any other oncolytic viruses, pembrolizumab, or any
other inhibitor
of PD-1, PD-Li or programmed cell death ligand 2 (PD-L2). Subjects should not
have a
history of evidence of symptomatic autoimmune diseases. Subjects should not
have active
herpetic skin lesions or prior complications of herpetic infection and should
not require
intermittent or chronic treatment with an anti-herpetic drug.
Talimogene ktherparepvec /placebo treatment
[0313] The first cycle of talimogene laherparepvec or placebo is 21 (+3) days.
Subsequent
cycles should be given every 2 weeks ( 3) days until week 9 and every 3 weeks
( 3) days
thereafter. On day 1 of cycle 1, the first dose of talimogene laherparepvec is
up to 4.0 mL of
106 PFU/mL or placebo. The second injection of up to 4.0 mL of 108 PFU/mL
should be
administered 21 (+3) days after the initial injection (i.e., no sooner than
day 22 but should not
be delayed more than 3 days after the 21-day time point). Talimogene
laherparepvec/placebo
should be administered until disappearance of injectable lesions, complete
response,
documented confirmed progressive disease by iRC-RECIST, intolerance of study
treatment,
24 months from the date of the first talimogene laherparepvec/placebo, or end
of study,
whichever occurs first. The treatment cycle interval may be increased due to
toxicity. When
talimogene laherparepvec or placebo injections and pembrolizumab are
administered on the
same day, talimogene laherparepvec or placebo should be administered first, if
possible.
Pembrolizumab treatment
[0314] Pembrolizumab at a dose of 200 mg is administered intravenously every 3
weeks ( 3
days). The second dose of pembrolizumab is administered 21 (+3) days after the
initial dose.
The treatment cycle interval may be increased due to toxicity. When talimogene
laherparepvec and pembrolizumab are administered on the same day, talimogene
laherparepvec should be administered first, if possible. Pembrolizumab dosing
is continued
until confirmed Progressive Disease (PD) per irRC-RECIST, intolerance to
treatment, 24
months from the date of the first dose of pembrolizumab, or the end of study,
whichever
occurs first. Discontinuation of treatment may be considered for subjects who
have attained a
confirmed Complete Response (CR) that have been treated for at least 24 weeks
with
pembrolizumab and had at least 2 treatments with pembrolizumab beyond the date
when the
initial CR was declared.
88

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
Example 10. Phase lb Clinical Trial Combining Talirnogene Laherparepvec and
Pembrolizumah In Patients with Recurrent or Metastatic Squainous Cell
Carcinoma of
the Head and Neck
[0315] A Phase lb trial (MASTERKEY232; ClinicalTrials.gov Identifier:
NCT02626000)
was designed in patients with recurrent or metastatic squamous cell carcinoma
of the head
and neck (SCCHN) to receive combination therapy of intralesional injection of
talimogene
laherparepvec with systemic administration of pembrolizumab. The primary
objective was to
assess the safety of this combination and secondary objectives included immune-
related
tumor responses.
[0316] Eligible patients, 18 years or older, had histologically confirmed
metastatic or
recurrent SCCHN of the oral cavity, oropharynx, hypopharynx, or larynx, and
the disease
was considered unresectable and not amenable to curative radiotherapy, and
measurable
disease suitable for intralesional injection (> 1 cutaneous, subcutaneous, or
nodal SCCHN
tumor > 10 mm, in longest diameter, either alone or in aggregate). Eligible
patients had
disease progression or recurrence after treatment with a platinum-containing
therapy.
Patients were required to have adequate performance status and hematologic,
renal, hepatic,
and coagulation function. Patients were excluded if they had active CNS
metastases and/or
carcinomatous meningitis, primary nasopharyngeal carcinoma, or were at risk of
a
compromised airway due to swelling/inflammation post-injection of the tumor,
previously
received talimogene laherparepvec, pembrolizumab, or other anti-PD-1 therapy.
All patients
provided written informed consent. Study procedures were approved by an
institutional
ethics committee at each site.
Study Design
[0317] This phase lb study was an open label, multicenter, single arm study
that primarily
evaluated the safety of intralesional talimogene laherparepvec in combination
with
intravenous pembrolizumab. Thirty-six patients received talimogene
laherparepvec and
pembrolizumab, of which the first 16 patients were part of the DLT analysis.
[0318] The first dose of intralesional talimogene laherparepvec, up to 8.0 mL
of 106
PFU/mL, was administered on day 1, week 1. Following doses, up to 8.0 mL of
108
PFU/mL, were given every 3 weeks. Up to 4 mL (total volume) of talimogene
laherparepvec
could be administered by intralesional injection at each treatment visit. The
volume delivered
to each injected lesion was contingent on the diameter of the lesion. The
injected volume per
lesion ranged from 0.1 mL for lesions <0.5 cm to 4.0 mL for lesions > 5 cm in
longest
89

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
diameter. Talimogene laherparepvec administration continued until
disappearance of
injectable lesions, complete response (CR), confirmed disease progression (PD)
per immune-
related response evaluation criteria in solid tumors (iRECIST), treatment
intolerance, 24
months from the first dose of pembrolizumab, or end of study, whichever
occurred first.
Pembrolizumab (200 mg) was administered intravenously ever 3 weeks beginning
on day 1,
week 1 (following the initial dose of talimogene laherparepvec on the same
day).
Pembrolizumab treatment was to be continued until iCR was achieved, confirmed
iPD,
intolerance to treatment, 24 months from the date of the first dose of
pembrolizumab, or end
of the study, whichever occurred first. The primary endpoint was the patient
incidence of
dose limiting toxicities (DLTs) starting from when both agents were given in
combination.
Incidence of DLTs in the first 16 evaluable patients and additional safety
data was reviewed
by a dose level review team (DLRT). The DLT evaluation period was 6 weeks from
the
initial dosing of both study treatments. To be considered in the DLT
evaluation, patients
needed to receive 2 doses of talimogene laherparepvec and 2 doses of
pembrolizumab in
combination or had a DLT after at least 1 dose of talimogene laherparepvec and
pembrolizumab in combination. The secondary endpoints included the following
immune-
related tumor responses per the investigator using irRECTST; objective
response rate (iORR;
complete response + partial response), complete response rate (iCRR), best
overall response
(iBOR), duration of response (iDOR), disease control rate (iDCR), and
progression free
survival (iPFS).
[0319] The primary analysis for the phase lb clinical trial was the tumor
response assessment
which was completed when the last subject enrolled had the opportunity to
complete the 9-
week response assessment.
Baseline Demographics and Characteristics
[0320] In the phase lb clinical trial, 36 patients with recurrent or
metastatic squamous cell
carcinoma of the head and neck were administered intratu moral talimogene
laherparepvec
and intravenous pembrolizumab. The baseline demographics and clinical
characteristics are
listed in Table 5.

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
[0321] Table 5. Baseline Demographics and Clinical Characteristics.
*I'alimogene Laherparepvec +
Pembrolizumab (N=36)
Sex ¨ n(%)
Male 29 (80.6)
Female 7(19.4)
Median (range) age, y 62 (35-77)
ECOG performance status, n(%)
9(25)
1 27(75)
Baseline HSV Status, n (%)
Negative 5 (13.9)
Positive 22 (61.1)
Unknown 9 (25.0)
Primary tumor site, n(%)
Oropharynx 9 (25.0)
Larynx 4(11.1)
Oral Cavity 20 (55.6)
Hypopharynx 3 (8.3)
PD-Ll status - n (%)
Positive 28 (77.8)
Not positive 3 (8.3)
Indeterminate 3 (8.3)
Missing 2 (5.6)
Dose Limiting Toxicity, Safety and Tolerability of Combining Talimogene
Laherparepvec
with Pembrolizumab
[0322] Sixteen patients were included in the dose limiting toxicity phase of
the lb trial. Of
these 16 patients, one (6.3%) DLT was observed, a grade 5 (fatal) arterial
hemorrhage. This
DLT rate of 6.3% supported the enrollment of an additional 20 patients in the
phase lb part
of the trial.
[0323] Thirty-six patients in the phase lb trial received at least one dose of
intratumoral
talimogene laherparepvec and at least one dose of intravenous pembrolizumab in
combination. There were no unexpected safety findings and the adverse events
were
consistent with those observed with the single talimogene laherparepvec and
pembrolizumab
montherapies. Grade 3 or higher treatment emergent adverse events were
reported in 5
(13.9%) patients considered related to talimogene laherparepvec and 3 (8.3%)
of patients
related to pembrolizumab. Of these grade 3 or highter treatment emergent
adverse events, 2
(5.6%) led to discontinuation of talimogene laherparepvec and 1 (2.8%) led to
the
discontinuation of pembrolizumab. Seven deaths were reported during the study
including
the DLT patient (arterial hemorrhage) which was considered related to
talimogene
91

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
laherparepvec and zero deaths were considered related to pembrolizumab. The
most
common treatment emergent adverse events were pyrexia (36.1%), dyspnea
(33.3%), and
fatigue (25.0%).
Anti-tumor Activity with Combined Talimogene Laherparepvec and Pembrolizumab
[0324] The primary analysis of the phase lb trial was triggered when the last
subject enrolled
had the opportunity to complete the 9-week response assessment. The primary
efficacy
endpoints were objective response rate (iORR), complete response rate (iCRR),
best overall
response (iBOR), duration of response (iDOR), and disease control rate (iDCR)
(response
evaluation by investigator using immune-related response evaluation criteria
in solid tumors
EirRECISTD. Per irRECIST, observations of iCR, il3R, and iPD require a
confirmatory scan
no less than 28 days from the original observation of the response in order to
be considered
confirmed in the response analysis.
[0325] Of the 36 enrolled patients, 28 (77.8%) had confirmed PD-L1 positive
tumors, 5
(13.9%) were HPV positive, and 13 (36.1%) were HPV negative, with 18 (50%)
unknown
(Table 6). The unconfirmed objective response rate (complete and partial
responses) per
iRECIST was 16.7% (95% CI, 6.4%- 32.8%) with a confirmed iORR of 11.1% (95%
CI,
3.1%-26.1%). The unconfirmed and confirmed disease control rate (complete,
partial, and
stable disease) per iRECIST was 38.9% (95% CI, 23.1%-56.5%). Long term follow-
up was
still ongoing at the time of the primary analysis.
[0326] Table 6: Best Overall Response.
Unconfirmed Confirmed
(N=36) =
Best Overall Response (iBOR) (136)
iORR (iCR/iPR) 6(16.7) 4(11.1)
95% CIb (6.4, 32.8) (3.1, 26.1)
iCR 0(0.0) 0(0.0)
iPR 6(16.7) 4(11.1)
iSD 8(22.2) 10 (27.8)
iPD 11 (30.6) 6(16.7)
iUE 1(2.8) 6(16.7)
ND 10 (27.8) 10 (27.8)

CA 03057157 2019-09-18
WO 2018/201028
PCT/US2018/029915
iDCR OCR/iPRASD) 14 (38.9) 14 (38.9)
95% Clb (23.1,56.5) (23.1, 56.5)
[0327] aPer irRECIST, observations of iCR, iPR, and IT'D require a
confirmatory scan no less
than 28 days from the original observation of the response in order to be
considered
confirmed in the response analysis.
[0328] bBinomial proportion with exact 95% confidence interval (CI).
93

Representative Drawing

Sorry, the representative drawing for patent document number 3057157 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Deemed Abandoned - Failure to Respond to an Examiner's Requisition 2024-07-26
Letter Sent 2024-04-29
Examiner's Report 2024-01-17
Inactive: Report - No QC 2024-01-16
Letter Sent 2022-11-25
Request for Examination Requirements Determined Compliant 2022-09-26
All Requirements for Examination Determined Compliant 2022-09-26
Request for Examination Received 2022-09-26
Common Representative Appointed 2020-11-08
Inactive: COVID 19 - Deadline extended 2020-03-29
Common Representative Appointed 2019-10-30
Common Representative Appointed 2019-10-30
Inactive: Notice - National entry - No RFE 2019-10-10
Inactive: Cover page published 2019-10-10
Application Received - PCT 2019-10-03
Inactive: IPC assigned 2019-10-03
Inactive: IPC assigned 2019-10-03
Inactive: IPC assigned 2019-10-03
Inactive: IPC assigned 2019-10-03
Inactive: IPC assigned 2019-10-03
Inactive: First IPC assigned 2019-10-03
National Entry Requirements Determined Compliant 2019-09-18
BSL Verified - No Defects 2019-09-18
Inactive: Sequence listing - Received 2019-09-18
Application Published (Open to Public Inspection) 2018-11-01

Abandonment History

Abandonment Date Reason Reinstatement Date
2024-07-26

Maintenance Fee

The last payment was received on 2023-04-05

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
Basic national fee - standard 2019-09-18
MF (application, 2nd anniv.) - standard 02 2020-04-27 2020-04-08
MF (application, 3rd anniv.) - standard 03 2021-04-27 2021-04-07
MF (application, 4th anniv.) - standard 04 2022-04-27 2022-04-07
Request for examination - standard 2023-04-27 2022-09-26
MF (application, 5th anniv.) - standard 05 2023-04-27 2023-04-05
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
MERCK SHARP & DOHME CORP.
AMGEN INC.
Past Owners on Record
ABRAHAM ANTONIO ANDERSON
JENNIFER LORRAINE GANSERT
KEVIN GORSKI
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2019-09-18 93 7,822
Drawings 2019-09-18 29 1,796
Claims 2019-09-18 8 442
Abstract 2019-09-18 1 61
Cover Page 2019-10-10 1 33
Examiner requisition 2024-01-17 4 205
Commissioner's Notice - Maintenance Fee for a Patent Application Not Paid 2024-06-10 1 543
Notice of National Entry 2019-10-10 1 202
Courtesy - Acknowledgement of Request for Examination 2022-11-25 1 431
International search report 2019-09-18 5 178
National entry request 2019-09-18 3 84
Request for examination 2022-09-26 3 65
Maintenance fee payment 2023-04-05 1 27

Biological Sequence Listings

Choose a BSL submission then click the "Download BSL" button to download the file.

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.

Please note that files with extensions .pep and .seq that were created by CIPO as working files might be incomplete and are not to be considered official communication.

BSL Files

To view selected files, please enter reCAPTCHA code :